v
Search
Advanced Search

Publications > Journals > Oncology Advances > Article Full Text

  • OPEN ACCESS

Natural Killer Cell Cellular-based Therapeutic Options to Manage Acute Myeloid Leukemia: Prospects and Challenges

  • Ogochukwu O. Izuegbuna* 
 Author information
Oncology Advances   2024

doi: 10.14218/OnA.2024.00013

Abstract

Over the past decade, significant progress has been made in managing acute myeloid leukemia (AML). However, refractory disease and relapse continue to pose major challenges. These issues highlight the need for innovative therapeutic options to achieve deeper remission and effectively treat refractory and relapsed diseases, thereby improving survival rates. Natural killer (NK) cell-based therapies have emerged as a promising option. NK cells, a specialized population of innate lymphoid cells, exhibit inherent anti-viral and anti-cancer capabilities. Unlike T cells, NK cells do not require prior antigen sensitization to eliminate their target cells, enhancing their potential as immunotherapeutic agents. However, NK cells often exhibit dysfunction in patients with hematological malignancies. Revitalizing these cells represent another immunotherapeutic strategy. Various NK cell-based therapies have been explored in recent decades, particularly in managing AML. These therapies include chimeric antigen receptor-NK cell therapy, bispecific and trispecific NK cell engagers (bi-specific killer cell engager (BiKEs) and tri-specific killer cell engager (TriKEs), and cytokine-induced memory-like NK cells. These therapies are also associated with fewer adverse events, such as neurotoxicity. Despite their potential for clinical cancer management, challenges such as the in vivo expansion of NK cells remain unresolved. This review summarizes the biology of NK cells and the diverse NK cell-based therapies being developed for the potential management of AML, as evidenced in preclinical studies and clinical trials.

Keywords

Natural killer cells, Acute myeloid leukemia, Cellular therapy, Cytokines, Chimeric antigen receptors natural killer (CAR-NK) cell therapy, Innate lymphoid cell

Introduction

Acute myeloid leukemia (AML) is a heterogeneous, clonal malignancy of myelogenous cells characterized by the accumulation of myeloid blast cells, primarily in the bone marrow, resulting in impaired production of normal blood cells. Until recently, the primary treatment option for AML was often cytotoxic chemotherapy. However, in 2017, midostaurin became the first targeted therapy approved for AML treatment in nearly four decades,1 followed by subsequent new approvals.2 With an increasing lifespan worldwide, the incidence of AML is also rising. According to Surveillance, Epidemiology, and End Results (SEER) statistics, in the United States alone, in 2022, there were an estimated 20,050 new cases of AML, representing 1% of all new cancer cases, and 11,540 deaths, representing 1.9% of all cancer deaths.3 This represents approximately a 6% increase in deaths compared to SEER data from 2019. Despite advancements in drug therapy, relapse remains a significant issue. Currently, hematopoietic stem cell transplant (HSCT) may be the only curative therapy available for AML, albeit with considerable risks and side effects. Therefore, there is a critical need for newer therapeutic options that can achieve greater complete remission (CR) with negative minimal residual disease. Immunotherapy represents one such promising option (Fig. 1).4,5

Timeline of FDA-approved medicines for acute myeloid leukemia (AML).
Fig. 1  Timeline of FDA-approved medicines for acute myeloid leukemia (AML).

CAR-NK, chimeric antigen receptors natural killer; CAR-T, chimeric antigen receptors T; CC-486, oral azacitidine; CIML, cytokine-induced memory-like; CPX-351, liposomal duanorubicin and cytarabine; FDA, Food and Drug Administration.

Immunotherapy in cancer is a type of treatment that harnesses the specificity and killing mechanisms of the immune system to target and eradicate malignant cells. Immunotherapy has been noted as a viable treatment strategy in managing various cancers and was voted “breakthrough of the year” by Science in 2013.6 Immune cells and biochemicals of the immune system are constitutively made to fight disease-causing microbes and their infected cells, as well as cells with the potential for neoplastic transformation. However, malignant cells have devised various means of evading immune cells through the loss of immunogenicity, upregulation of negative regulatory pathways, or creating an immunosuppressive microenvironment, thereby making immune cells less potent in destroying cancer cells.7,8 Extensive research in cancer immunotherapy and the dynamic interactions between cancer cells and host immune cells have brought up innovative ways of boosting the host immune cells or initiating novel ways to elicit immune responses in fighting cancer, which has led to the approval of new therapies against both solid and hematological cancers.9,10 Recently, Natural killer (NK) cell cellular-based therapies have been muted as one of the novel strategies in fighting cancers, especially hematological cancers. It has been observed that there is a quantitative and qualitative dysfunction in NK cells in hematological cancers. There is further impairment in their numbers and function as a result of chemotherapy and radiation used during treatment.11,12 Restoration of these immune impairments can improve therapeutic outcomes. Over the years, a better understanding of NK cell immunobiology coupled with improvements in molecular biology techniques have led to increased development in the field of NK cell cellular-based therapy in hematological cancers, including chimeric antigen receptor (CAR)-modified NK cells,13–17 adoptive cell transfer,18–20 cytokines,21,22 bispecific natural killer cell engager (BiKE),23–25 drug treatment, etc.26–28 Despite the major developments in NK cell-based therapies, especially in AML, it is yet to make inroads into the clinics. This is a result of different factors, the chief one being that clinical trials are still in progress in many of them. In this work, I look into the biology of NK cells, the various NK cell-based therapies being developed in preclinical and clinical trials, and the challenges faced getting them to the clinic.

NK cell biology

NK cells are a distinct group of innate lymphoid cells capable of identifying and destroying virally infected and tumor cells. They can be classified based on CD56 (neural cell adhesion molecule) and CD16 expression. NK cells constitute 5–20% of circulating lymphocytes in humans.29 There are two primary subsets of NK cellsCD56bright or CD56dim. Approximately 90% of circulating NK cells are CD56dim, representing the final stage of NK cell maturation. This subset expresses killer cell immunoglobulin-like receptors (KIRs), which are inhibitory receptors and cytotoxic effector proteins such as perforin and granzyme B at rest. There is also an increased expression of CD16 (FcγRIIIa), which plays a role in targeting antibody-opsonized cells. The remaining 10% of the NK cell population are CD56bright and express lower levels of cytotoxic effector proteins at rest. CD16 is also expressed at lower levels in this subset. Unlike the CD56dim subset, which expresses KIRs as an inhibitory molecule, CD56bright NK cells express CD94/NKG2A, CD94/NKG2C, and NKG2D receptors. Compared to the CD56dim subset, bright NK cells possess specialized chemokine and homing receptors such as CCR7.30 Additionally, the CD56 bright subsets can produce immunoregulatory cytokines such as interferon (IFN)-γ, tumor necrosis factor (TNF)-α/β, and interleukin (IL)-10 upon combined cytokine receptor stimulation.31–33 Traditionally, CD56bright NK cells exhibit low antitumor activity at rest, unlike the CD56dim subset known for its robust cytolytic activity. However, CD56bright NK cells from multiple myeloma (MM) patients have enhanced ex vivo functional responses when primed with IL-15.34,35 NK cells mature in the bone marrow (BM) and other secondary lymphoid tissues; however, the BM is the primary site for NK precursor cells, and unlike T cells,36–38 other secondary sites such as the spleen and thymus do not hinder NK cell growth and function.36,37,39 Further stages of NK cell ontogeny occur in secondary lymphoid tissues such as the liver, lymph nodes, and tonsils.40,41 Specifically, in the parafollicular T cell region of the lymph node, which is rich in CD56bright NK cells, differentiation into mature CD56dim NK cells occurs after stimulation by IL-2.42

The Lin−CD34+CD133+CD244+ HSCs are known to differentiate into CD45RA+ lymphoid-primed multipotential progenitor (LMPP) in the early stages of development, which are also found to be CD38- and CD10- but have CD62L.43,44 The LMPPs can differentiate into multiple lymphoid lineages and some residual myeloid lineage but lack erythroid and megakaryocytoid potentials and no self-renewal capacity.45 The LMPP transit into the common lymphoid progenitor (CLP), which lacks the potential for myeloid differentiation but can make lineage commitment into all subsets of lymphocytes, i.e., Pro-B, Pre-T, NK progenitors (NKPs), or other innate lymphoid cells (ILCs).46 CLP were earlier thought to be Lin− cKitlowSca-1lowCD127hi (IL-7Rαhi) but were later refined to include high expression of Fms-related tyrosine kinase 3 (Flt3).47 CLPs have also been discovered to be associated with the expression of Ly6D. This surface marker divides CLPs into two distinct populations. The Ly6d− subset of CLP, called all lymphoid progenitor, has T and NK potentials, whereas the Ly6d+ subset, called BLP (B-cell-biased lymphoid progenitor), up-regulates the B-cell-specifying factors Ebf1 and Pax5, thus acting as B cell progenitors.48 It should be noted that NK cells were for some time the only known ILCs - innate lymphocytes that cannot express RAG-dependent rearranged antigen-specific cell surface receptors until another innate lymphoid cell known as the lymphoid tissue-inducer cell was discovered in the 1990s and subsequently helper-like innate lymphoid lineages from 2008. ILCs are classified into five groups, and this is based on their developmental course and cytokine profile. They include the cytotoxic NK cell, lymphoid tissue-inducer cells which express the integrin α4β7, lymphotoxin (LT) α1β2, and lymphoid cytokine receptors, and helper-like ILCs (ILC1, ILC2, and ILC3) with their distinct functional expression like CD4+ T helper (Th) type 1, Th2 and Th17 cells.49–52 The families of innate lymphocytes share a common progenitor known as the early innate lymphoid progenitor; the cytokine-producing ILCs also have a more restricted progenitor known as common helper-like innate lymphoid cell progenitor.53–55 All ILCs except NK cells require GATA-3 for their differentiation.56 In addition, NK cells and ILC1 cells depend on two evolutionary-related T-box transcription factors (TFs): eomesodermin (EOMES). T-box expressed in T cells (T-bet) for their development. However, EOMES is strictly required to develop NK cells, while ILC cells do not develop without T-bet in conjunction with Aiolos and Bcl6.55,57–59GATA-3, B-cell lymphoma/leukaemia 11B (BCL11B) and RAR-related orphan receptor alpha (RORα) are required for the development of ILC2 cells as well as the control of the production of type 2 effector cytokines, IL-5, IL-13, and IL-4.60,61 The group 3 ILC cells depend on GATA-3, RAR-related orphan nuclear receptor γt (RORγt), and Hypoxia-Inducible Factor (HIF-1) to develop and produce cytokines IL-17 and IL-22.59,62 In all these, mature ILCs can be generated from CLPs.55

While the ontogeny of NK cells is not fully understood, their development has been classified into six stages. Stage 1 begins with CLPs transitioning into NKPs characterized by the expression of CD7+, CD127+ (IL-7Rα+), CD122+ (IL-2Rβ+), CD117+ (c-Kit+), and IL-1R1low. The acquisition of CD122 indicates a commitment to the NK lineage, promoting NK cell differentiation, functional maturation, and survival.63–65 Stage 2a of pre-NK cells is defined by CD3ε−CD7+CD127+ cells, while the transition from stage 2a to 2b is marked by the expression of IL-1R, a receptor for IL-1β at stage 2b.66 The progression from stage 2b pre-NK cells to stage 3 immature NK cells is indicated by the expression of activating receptors CD335 (natural cytotoxicity receptor, NCR1, NKp46), CD337 (NCR3, NKp30), and CD161.67 NKG2D, which uses the DAP10 adaptor protein, along with NCR1 and NCR3, which utilize CD3ζ and FcεRγ, respectively, characterize this development.

Stage 4 of NK cell development is subdivided into stages 4a and 4b, with stage 4a being NKp80 negative and characterized by increased levels of NKG2D, CD335, CD337, inhibitory NKG2A (CD159a, containing two immunoreceptor-based tyrosine inhibitory motifs (ITIMs) and CD161 (NK1.1, KLRB1, NKR-P1A) which are CD56bright. The Stage 4b shows positivity for NKp80 while maintaining their CD56bright status. Stage 5 is characterized by the transition from CD56bright to CD56dim, which are mature NK cells. This stage involves the gradual up-regulation of CD94/NKG2C and CD16 (FcγRIII), and down-regulation of CD56, c-Kit (CD117), and CD94/NKG2A.68 CD56bright NK cells, considered less mature, are primarily found in secondary lymphoid tissues, unlike the CD56dim that form the majority of NK cells in circulation.12,69

Ultimately, the terminal maturation of CD56dim NK cells is marked by the expression of CD57 (HNK-1, Leu-7) and killer cell immunoglobulin-like receptors (KIR+/CD158+), which constitutes stage 6 of NK cell development (Fig. 2, Table 1).70–72

Schematic diagram of NK cell development from the hematopoietic stem cell (HSC) to the terminal stage 6.
Fig. 2  Schematic diagram of NK cell development from the hematopoietic stem cell (HSC) to the terminal stage 6.

CLP, common lymphoid progenitor; NK, natural killer; NKP, NK progenitor.

Table 1

Surface antigens expression at different stages of NK cell development

Surface markersStage 1Stage 2aStage 2bStage 3Stage 4aStage 4bStage 5Stage 6
CD34+++
CD10++/−+/−
HLA-DR+++
CD117++++
CD127++++
CD122++++++
CD161−/+++++
CD56++++++
CD94+++/−+/−
NKG2A++
NKG2D++++
NKp30++++
NKp46++++++
NKp80+++
NKG2C++
CD16++
KIRs++
CD57+

NK cells recognition of self from non-self

The process of NK recognition of self from non-self is still being debated. This education process results from NK cells interaction with self-major histocompatibility complex (MHC)-I.73 Several studies examining the basis of tolerance in NK cells have linked it to MHC-I surface expression. The KIR family of receptors is the NK group of receptors that primarily associate with MHC-1. Through them, MHC-I regulates NK cell function. In trying to explain the NK cell education process, Yokoyama and colleagues brought up a theory called the NK cell licensing hypothesis, which states that for NK cells to respond to subsequent stimuli received by inhibitory receptors, they must first engage in self-MHC class I. This is termed “licensing”. Conversely, the NK cells that could not engage in self-MHC class I were considered “unlicensed”.74,75 Thus, this process gives rise to two kinds of self-tolerant NK cells which are: (a) the licensed NK cells, which are capable of maintaining self-tolerance by direct inhibition through binding to self-MHC and (b) the unlicensed NK cells, which cannot engage self-MHC but are self-tolerant due to their inherent resistance to stimulation received through activating receptors.

Later, Raulet and Vance introduced their NK cell self-tolerance model, termed the arming and disarming model. According to the arming model of NK cell education, the KIR inhibitory receptor interaction with MHC class I molecules gives rise to inhibitory signals that promote functional maturation of human precursor NK cells but not mature NK cells. This hypothesis appears counterintuitive in that these receptors are essentially inhibitory. However, signaling through these receptors may seem more complicated than previously thought. On the other hand, the disarming model proposes that precursor and mature NK cells that lack self-MHC-I inhibitory receptors are rendered hyporesponsive upon receiving sustained positive signaling via activating receptors.76 Thus, these models show that increased expression of inhibitory receptor signaling in comparison to activating signaling invariably leads to a heightened response of NK cells; therefore, NK cells with functional copies of KIR genes are functionally more competent than those without in their education process.77 As a result of the alterations in the expression of inhibitory receptors during NK cell development, various combinations of inhibitory receptors can be expressed on distinct NK cells, particularly in a disease state, thus making it function like a rheostat to set a quantitative threshold of NK cell responsiveness during the education process.78 This is the rheostat model, which incorporates concepts from the licensing and disarming model that different inhibitory receptors can bind MHC ligands with varying affinities, and the interactions between the various inhibitory receptors and the expressed MHC molecules will result in varying degrees of inhibition between distinct NK cells which allows for a range of NK cells responses.79,80 Just like the diversity of the MHC molecules, the KIR displays a high level of polymorphism. The KIR haplotypes are grouped into two primary sets: “A” and “B”.81 The KIR A haplotypes mainly contain inhibitory KIR genes and only one activating KIR gene, KIR2DS4. On the other hand, KIR B haplotypes have different numbers and combinations of activating KIR genes besides inhibitory KIR genes.

NK cell signaling and effector functions

Unlike T cells, NK cells do not express clonotypic receptors. Nevertheless, they can still generate significant anti-tumor cytotoxicity and produce inflammatory cytokines. These functions are regulated by an array of germline-encoded activating and inhibitory receptors, including NKG2D, NCR1, NCR2, NCR3, NKG2C, CD244, Ly49D, Ly49H, KIRs, CD94/NKG2A, and leukocyte Ig-like receptor 1 (LIR1). These receptors are transmembrane proteins with an extracellular ligand-binding portion and an intracellular cytoplasmic tail. The cytoplasmic tail of inhibitory receptors contains immunoreceptor tyrosine-based inhibitory motifs, which can directly activate their protein phosphatases. Conversely, activating receptors, which lack signaling domains in their cytoplasmic tails, indirectly stimulate protein kinases by recruiting adaptor proteins containing immunoreceptor tyrosine-based activation motifs (ITAMs). The adaptor molecules propagating activation receptor signaling include FcεRIγ, CD3ζ, and DAP12.

NKG2D and Ly49H can also propagate signals through the Tyr-Ile-Asn-Met (YINM) motif present within the adaptor, DAP10. The activating receptor NKG2D is a type II transmembrane and C-type lectin-like type II homodimeric receptor that is involved in NK cell lysis (just like other activating receptors, (NCR) NKp46 (NCR1), NKp30 (NCR3), and NKp44 (NCR2)). It is constitutively expressed on NK cells and mediates signaling through the adapter proteins DAP10 and DAP12 via YINM and ITAM tyrosine-based signaling motifs- DAP10 is involved in the recruitment and activation of the p85α subunit of PI(3)K and Grb2, while DAP12 is involved in the recruitment of ZAP70 and Syk to initiate NKG2D-mediated NK cell activation.

In addition to these activating receptors, co-receptors such as 2B4, NTB-A, DNAM-1, CD59, and NKp80 play complementary and synergistic roles in NK cell activation. 2B4 and NTB-A, part of the signaling lymphocyte activation molecule (SLAM) family, enhance the potentiation and cytotoxic activity of NK cells triggered by primary receptors.82 These two co-receptors, associated with the SLAM-associated protein (SAP), a molecule involved in X-linked lymphoproliferative syndrome type 1 (XLP-1) — a severe form of immunodeficiency83,84 — have been noted to deliver inhibitory signals in the absence of SAP, rather than activating signals.84–86

CD59, a glycosylphosphatidylinositol (GPI)-linked protein, and a paroxysmal nocturnal hemoglobinuria marker depends on the simultaneous engagement of NKp46 and NKp30 receptors via the tyrosine phosphorylation of CD3zeta chains to enhance NK cell-mediated cytotoxic activity.87 Low CD59 is associated with increased proliferation and abnormal coagulation function in AML.88 An adhesion molecule, DNAX Accessory Molecule (DNAM-1 or CD226), is involved in NK cell activation. DNAM-1 has two ligands, poliovirus receptor (PVR) and Nectin-2, widely expressed in hematological cancers.89 The dual interaction of the ligands with the activating coreceptor, DNAM-1, and the inhibitory receptors CD96 and T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) (DNAM-1 enhances NK cell-mediated cytotoxicity via PVR and Nectin-2, whereas TIGIT interaction with these ligands leads to a reduction in IFN-γ production by NK cells, as well as a diminished NK cell-mediated cytotoxicity) makes them an ideal target for immunomodulation in cancer.90–93 Studies have shown a reduced expression of DNAM-1 in AML while the inhibitory receptors TIGIT and T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) are increased.94 Consequently, loss of DNAM-1 and reduced expression of PVR is a primary NK cell escape mechanism in AML.95

Human leukocyte antigens (HLA) class I and non-classical MHC class Ib molecules such as HLA-E are recognized by the inhibitory receptors KIRs and CD94/NKG2A. KIRs are clonally distributed; only a fraction of NK cells express a given KIR, making them highly polymorphic. The HLA class I molecules that express either the Bw4, C1, or C2 motifs are the ideal ligands for KIR. On the other hand, HLA-C alleles that are characterized by Lys at position 80 (HLA-CLys80) are recognized by KIR2DL1, while KIR2DL2/3 recognize HLA-C alleles characterized by Asn at position 80 (HLA-CAsn80). Likewise, KIR3DL1 is specific for HLA-B alleles sharing the Bw4 supertypes specificity (HLA-BBw4), and KIR3DL2 recognizes HLA-A3 and -A11 alleles.89 The KIR system acts through specific interactions and varying degrees of signal strength to diversify NK cell stimulation. Thus, weakly inhibitory KIR/HLA combinations permit a lower threshold for cell activation and vice versa. Therefore, target cells are susceptible to NK-mediated killing when there are no effective inhibitory interactions. A study by Dai et al.96 showed that increased KIR2DL1, KIR2DL3, KIR2DL4, KIR3DL1, and KIR3DL2 mRNA levels were significantly related to poor prognosis and overall survival (OS) in AML patients (Fig. 2).

In performing their effector functions, i.e., cytotoxic death of the target, NK cells are reported to use various mechanisms. This requires specific processes. In target destruction, NK cells first recognize their target through specific molecular mechanisms. These inhibitory and activating receptors recognize surface molecules expressed at steady state and stress-induced molecules, respectively. Once a target cell is recognized, there is a direct interaction of the NK cell with the target through the formation of an immunological synapse, which facilitates target cell death through some mechanisms. Human NK cells kill their target primarily by releasing lytic granules in a process called ‘degranulation’.97 These lytic granules are delivered to the target cell through membrane fusion at the immunological synapse. This process involves cytoskeletal rearrangement, which includes actin polymerization and polarization of the cytoskeletal rearrangement-assisted microtubule-organizing center towards the target cell.98–101 Once polarized, These lytic granules move along the microtubules and at the immunological synapse fuse with the target cell membrane and release their lytic enzymes, which cause the activation of an apoptotic process within the target cell.102 The major components of the lytic granules in the “degranulation” process are Granzyme B and perforin. Perforin, a 60–70-kDa pore-forming glycoprotein, forms pores in target cells, leading to osmotic lysis. A partial deficiency in perforin production causes increased susceptibility to hematological cancers.103 On the other hand, Granzyme B, a class of serine proteases, can induce apoptotic cell death through caspase-dependent and independent mechanisms.104

Another mechanism through which NK cells eliminate their targets involves the engagement of death receptors on target cells via their cognate ligands, which are present on the NK cells.105 The TNF-related apoptosis-inducing ligand-receptor (TRAIL-R) and Fas (CD95) are two such death receptors activated by their respective ligands, Fas ligand (FasL, CD95L) and TRAIL. The binding of these receptors by their ligands induces a conformational change through receptor oligomerization and the recruitment of adapter proteins, initiating apoptosis either directly through effector caspases or indirectly via the intrinsic mitochondrial pathway.106,107

In addition to their cytotoxic capabilities, NK cells are potent producers of pro-inflammatory and immunosuppressive cytokines, primarily mediated by CD56bright NK cells, which are less cytolytic.31,32 The primary cytokines produced include IFN-γ and TNF-α, and, depending on the inflammatory environment, IL-5, IL-10, IL-13, and some growth factors like IL-3, G-CSF, and GM-CSF. NK cells also secrete chemokines such as CCL1, CCL2/MCP-1, CCL3 (MIP-1α), CCl4 (MIP-1β), CCL5 (RANTES), XCL1 (lymphotoxin), CXCL10/IP-10, and CXCL8 (IL-8), which attract effector lymphocytes and myeloid cells to inflamed tissues.108–110

Several transcriptional regulators are involved in the production of these cytokines, including the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), the c-Fos and c-Jun heterodimer of the AP-1 TF genes, and the nuclear factor of activated T cells.111–114 While NK cell cytokine secretion can be both beneficial and deleterious, NK cells have been observed to display “split anergy,” a phenomenon characterized by increased cytokine secretion but reduced cytotoxicity, particularly following interactions with cancer stem cells.109 This process is mediated by IFN-γ. However, some cytokines mediate beneficial immunoregulatory functions: IFN-γ facilitates dendritic cell maturation and indirectly promotes adaptive T-cell responses, activating helper T cells to a Th1 phenotype.115 Similarly, TNF-α is involved in B-cell proliferation and exhibits anti-proliferative effects on tumor cells. TNF-α also mediates endothelial activation, leading to increased production of adhesion molecules and inflammatory cytokines.

In addition to its MHC-I targeting of diseased cells, NK cell is also involved in antibody-dependent cellular cytotoxicity. NK cells possess Fc receptors that can bind antibodies in their Fc region. They bind to the Fc portion of immunoglobulins through their own FcγRIIC/CD32c and FcγRIIIA/CD16a.116,117 FcγRIIC has an ITAM in its cytoplasmic tail just as FcεRI-γ chains or CD3-ζ chains within the cell membrane equally have, though the primary activating receptor is the low-affinity FcγRIIIA/CD16a that binds the Fc domain of IgG. However, upon binding to FcγR there is phosphorylation of the ITAMs, which initiates a signal cascade, i.e., there is binding to tyrosine kinases ZAP-70 and Syk, with subsequent activation of the PI3K, NF-κb, and extra-cellular signal regulated kinases (ERK) pathways that cause NK cell degranulation, cytokine release, and tumor cell lysis.118–120

Although NK cells do not have clonotypic receptors like T cells, studies have shown that a relatively small population can elicit memory-like responses.121,122 Memory NK cells were first described in mice deficient in T & B cells. Following secondary exposure to specific haptens such as 2,4-dinitrofluorobenzene and oxazolone, there was a hypersensitivity response against the haptens mediated by NK cells on contact with these haptens. The sensitized NK cells that were adoptively transferred persisted for about four weeks.123 The development of memory NK cells has been studied in mice infected with murine cytomegalovirus (MCMV). The C57BL/6 mice were an ideal choice to study memory NK cells. This is because of their expression of the activating receptor Ly49H, which is specific for the viral glycoprotein m157 expressed on virally infected cells. Following MCMV infection, a small population of NK cells persists despite NK cell contraction. When isolated and adoptively transferred to naïve neonate mice, these memory NK cells that lack effective MCMV defense were better able to protect and prevent MCMV-mediated death compared to NK cells isolated from naïve hosts.121,124 More research has shown that cytokine-mediated activation, particularly IL12 and IL18, can induce NK cells with such memory traits, and when adoptively transferred back into mice, led to heightened IFN-γ secretion for some weeks along with cytotoxicity usually observed in resting NK cells.125,126 Similarly, Jin et al.126 showed that the in vivo pre-activation and re-stimulation of NK cells with interleukins (IL-12, IL-15, and IL-18) led to enhanced IFN-γ secretion which could be transferred to the next generation of NK cells and was associated with prolonged survival. The increased IFN-γ secretion was suggested to be likely NKG2D-dependent. Also, Brillantes and Beaulieu have shown that NK cells can produce memory and memory-like responses towards different microbial pathogens.127 The ability of these memory NK cells to produce enhanced levels of IFN-γ with cytotoxic granules and their ability to persist for a long time, these cells are being muted as potential cancer chemotherapies.128–130 It remains to see how they can be harnessed.

NK cell dysfunctions in AML

From its biology, NK cells can eliminate malignant cells by exerting both direct and indirect anti-neoplastic effects through their cytotoxic and immunoregulatory functions, which are essential for directing an enhanced immune response against cancer cells. However, studies have shown that in AML, the immune microenvironment is impaired, including myeloid and erythroid differentiation, macrophages and T-cell functions, osteogenesis, and NK cell immune surveillance.131–136 Using single-cell RNA sequencing, Guo et al.137 observed significant differences between normal and AML BM immune cells. Kutznesova et al.138 also reported impaired degranulation of NK cells in ex vivo AML models with increased transcriptional signatures observed in IL-6-STAT3 and IL-1β/TNFα. Thus, impaired immune function, particularly in NK cells, is one of the means that AML escapes immune surveillance. AML evade NK cell immune surveillance through different ways, including 1) the reduction in the number of NCRs on NK cell surface, 2) the overexpression of the inhibitory receptors KIRs and NKG2A with the resultant increase in inhibition of cytotoxicity, 3) interference with the maturation of NK cells with the majority of cells expressing CD56bright/dim KIRs- CD57- 4) the expression of checkpoint inhibitors like PD-1 and TIGIT resulting in NK cells with reduced ability to proliferate and lower cytotoxic and cytokine-producing capabilities.

One of the features of AML progression is the reduction in the number of functionally active NK cells. There is an inverse correlation between the anti-leukemic activity of NK cells and disease progression in AML with the observed suppression of NK cell number during active disease, increase in number in remission, and suppression again in the event of a relapse.139–141 Conversely, NK cell fusion post-HSCT was associated with reduced relapse and without an increased incidence of graft-versus-host-disease; in a study, the 1-year OS, CR rate, ORR, relapse rate (RR) of acute and chronic graft-versus-host disease (GvHD) rates were 69%, 42%, 77%, 28%, 24.9% and 3.7%, respectively.142–144 In addition, NK cells significantly correlate to OS and risk stratification in AML patients.145

The NCRs are surface receptors that are important in NK cell cytotoxicity. Blocking of these receptors inhibits NK cell cytotoxicity. NCR expression on NK cells is either bright (NCRbright) or dull (NCRdull), and most healthy individuals express the NCRbright phenotype. Studies have shown a correlation between NCR expression and NK cell-mediated cytotoxicity.146,147 In AML, these receptors are also under-expressed, affecting NK cell cytotoxicity and cytokine production.148,149 While there is a low expression of NCRs by NK cells in AML, the expression of ligands for NK cell activating/inhibitory receptors is also defective.137,150,151 This lack of expression of NCR ligands on their cell surface makes it difficult for NK cells to target them via NCR engagement, allowing them to escape immune surveillance. For example, the activating receptor found on NK cells, NKG2D interacts with its ligands (NKG2D-L), which comprise two members of the MHC class I-related chain (MIC) family (MICA, MICB) and six members of the UL16-binding protein (ULBP) family of proteins (ULBP1–6) and are generally not found on healthy cells but are induced on the surface of malignant cells. The NKG2D/NKG2D-L system has been observed as an important player in tumor development generally in cancer patients. The expression of some of these NKG2D-L is regulated by c-myc and DNA methylation, making them therapeutic targets for NK cell therapy.152–155 Furthermore, tumor cells cause proteolytic shedding by metalloproteases and release of soluble NKG2D-L, causing downregulation of NKG2D and blocking receptor activation.156 Likewise, the expression of NKG2D/NKG2D-L has been observed to decrease in the later stage of AML development, thus impairing NK cells destruction of AML cells. The absence of NKG2D-L in AML cells has also been noted to be responsible for disease relapse; in addition, increased DNA methylation for NKG2D-L is found in AML cells, which can be reversed with demethylating agents. In their investigation of soluble NKG2DL in 205 leukemia patients, Hilpert et al.156 discovered that about 75% expressed at least one NKG2DL at the surface. All investigated patients had elevated soluble NKG2DL levels in their sera. They also demonstrated that soluble NKG2DL in their sera reduced NKG2D expression in NK cells, which impaired antileukemic activity.157 Thus, AML cells escape NK cells’ target and elimination by reducing the levels of NKG2D-L expression. The clinical importance of the NKG2D/NKG2D-L system is also highlighted in a study that shows that the blocking of MICA/MICB shedding prevented cancer cell growth in immunocompetent mouse models and with the reduction of melanoma metastasis in a humanized model.158 In contrast, DNAM-1 and its ligands (CD112, 155) are frequently expressed in leukemic blasts, and its expression is associated with a favorable prognosis.149,159,160 However, under-expression of DNAM-1 has also been reported in AML, and it correlates with poor NK cell lysis.151,161,162

Alterations in the expression of inhibitory receptors have also been described in AML. In their research, Sandoval-Barrego et al.163 reported that patients with all FAB types of AML had overexpression of inhibitory receptors CD158b and NKG2A and decreased expression of the activating receptor NKp46. The CD94/NK group 2 member A (NKG2A) heterodimeric receptor binds to the non-classical HLA-E on cancer cells. It is one of the most prominent NK inhibitory receptors. NKG2A levels have been higher in the peripheral blood NK cells of patients with AML compared to NK cells of age-matched controls164. Its ligand, HLA-E, is known to be overexpressed in several cancer types, and it is also associated with poorer prognosis.164,165 The administration of a novel anti-human NKG2A antibody was able to impede tumor cell growth in leukemic cells, suggesting that HLA-E could be a therapeutic target.166 The KIR inhibitory receptors have also been studied. Shen et al.167 reported that inhibitory KIR ligands were present in significantly higher frequencies in the prognostically poor risk group than in those with favorable risk. Ghasemimehr et al.168 in their research of gene expression of activating and inhibitory receptors of NK cells in patients with newly diagnosed AML before and after induction therapy, reported a 6-fold increase in KIR2DL1 expression compared to healthy controls and a significant decrease in mRNA expressions of KIR2DL1 and NKG2A after induction therapy. Yang et al.169 also reported that the levels of other inhibitory receptors like TIM-3, ILT-4, ILT-5, and PD-1 were increased in NK cells from patients with AML (Fig. 3).

Schematic representation of NK cell activation and inhibitory receptors.
Fig. 3  Schematic representation of NK cell activation and inhibitory receptors.

CTLA-4, cytotoxic T-lymphocyte-associated antigen 4; DNAM-1, DNAX accessory molecule-1; KIR2DL1, killer cell immunoglobulin-like receptors two Ig domains and long cytoplasmic tail 1; KIR3DL1, killer cell immunoglobulin-like receptors three Ig domains and long cytoplasmic tail 1; LIR-1, leukocyte Ig-like receptor 1; NK, natural killer; NKG2D, natural killer group 2 member D; PD-1, programmed death-1; TIGIT, T cell immunoreceptor with Ig and ITIM domain; TIM-3, T-cell immunoglobulin and mucin-containing domain;

Defective maturation of NK cells

The NK cell development process involves different stages regulated by cytokines and transcription factors. The earlier described process moves them from the precursor stage through different maturation phases until they acquire full maturation with the expression of a host of receptors, especially the NKG2A or KIRs. This process transforms the NK cell into a cell with a high cytotoxic capacity that can recognize and eliminate cancer cells and viruses. However, this process can be hijacked by AML. Mundy-Bosse et al.,170 in their study on AML cells evasion of NK cells using specific murine maturation markers, showed that there was the selective loss of the intermediate (CD27+CD11b+) phenotype with the upregulation of the immature phenotype (CD27+CD11b−). The NK cells in AML also had lower levels of T-bet and EOMES along with the upregulation of microRNA miR-29b, a regulator of T-bet and EOMES, indicating a block in NK cell differentiation by AML. In their study on AML patients, Chretien et al. delineated three groups based on their NK cell maturation profile.171,172 This include: the hypomaturation (CD56bright/dim KIRs− CD57−), intermediate (CD56dim KIR−/+ CD57−/+) and hypermaturation (CD56dim KIRs+ CD57+) groups.171,172 They equally reported that patients in the hypomaturation group showed a poor 3-year overall survival and relapse-free survival, suggesting that maturation profiles of NK cells in AML may play an important role in prognostication and clinical course of the disease.171,172 In their most recent work (NCT02320656), Chretien et al.173 were able to demonstrate the presence of a moderate to increased number of CD56−CD16+ unconventional NK cells that showed a lower expression of NKG2A, as well as the activating receptors NKp30 and NKp46 in about a quarter of AML patients studied. These NK cells had a significantly decreased OS and event free survival (EFS) and a poor clinical outcome. Liu et al.,174 on their part, showed the expression characteristics of antigens and functional markers of NK cells in AML patients; NK cells were divided into two groups: CD3-CD56highCD16- (CD56high) and CD3-CD56dimCD16+ (CD56dim). The expression of CD56high NK cells was higher in AML patients than in healthy controls, and DNAM-1 expression was significantly low in CD56high NK cells, while NKG2D, DNAM-1, and perforin were significantly low in CD56dim NK cells.174 Single-cell profiling also revealed three subsets of NK cells in the bone marrow of AML patients, which also showed stress-induced repression of NK cell effector functions. This also showed the role AML plays in NK maturation and how it affects the course of the disease.

Immune checkpoint inhibitor expression

Immune checkpoint molecules are part of the arsenals of the immune system that play an important role in self-tolerance and the prevention of lysis of self-cells. Immune checkpoint molecules are expressed on many immune cells.175,176 Some immune checkpoint molecules expressed in NK cells include PD-1, TIM-3, LAG-3, TIGIT, and SIGLEC-7. Mature NK cells are known to express PD-1 when stimulated by MHC class I-deficient tumor cells or infected cells. These cells display reduced proliferative and cytolytic abilities and lowered cytokine production. Targeting immune checkpoints has been clinically proven and approved for managing some cancers, and the inhibition of PD-1 interaction with its ligand PDL-1 has been shown to restore NK cytolytic activity in some cancers.177–179 PDL-1 expression is elevated in AML patients, though its clinical significance to NK cell function is not well understood.180 Elevated PD-L1 expression in AML is associated with poor OS rate.181–183

Another immune checkpoint protein, the TIM-3 originally described on T-cells, is known to be expressed on the surface of NK cells, while its ligand Galectin-9 is also expressed in AML blasts. TIM-3 is reported to be associated with disease progression in cancer. In AML, TIM-3 is reported to be associated with poor prognosis. However, there is contradictory evidence to this. Darwish et al.184 and Kamal et al.185 reported TIM-3 as a poor prognostic marker in AML, while Xu et al.186 and Rakova et al.187 reported it as an excellent prognostic marker. High levels of soluble Galectin-9 have been demonstrated in the serum of AML patients. Its interaction with TIM-3 on leukemic stem cells activates the NF-kB and β-catenin pathways, which play a role in leukemic cells’ self-renewal.188

NK cell-based immunotherapy in AML

Adoptive NK cell transfer

While T-cell immunotherapy has gained prominence and approval in managing hematological cancers, NK cells have shown great promise; moreover, alloreactivity of NK cells in the allo-HSCT setting, which is triggered by a mismatch between the inhibitory receptors on the donor NK cells and the HLA class I molecules on recipient cells has been observed and muted as a therapeutic strategy, especially in the management of leukaemia.189,190 This alloreactivity of NK cells in leukemia is known to be mediated through the graft-vs-leukemia effect. It is also beneficial in preventing GvHD by destroying the recipient’s antigen presenting cells and fighting some infections. Ruggeri et al.,191 in their study of the impact of donor-versus-recipient NK cell alloreactivity on survival in acute leukemia patients, reported EFS at five years of 60% in those with the KIR ligand incompatibility versus 5% in those without the KIR ligand incompatibility. The KIR ligand incompatibility was the only independent predictor of survival in AML.191 In a related study, Mancusi et al.192 demonstrated the effect of the KIR ligand–mismatched NK cell donors on acute leukemia. They showed that in 69 patients that underwent HSCT with donor-vs-recipient NK-cell alloreactivity, there was a reduced risk of non-relapse mortality, superior EFS, and a 50% reduction in infection rate when the transplant was from donors with KIR2DL1 and/or KIR3DL1.192 Taken together, the adoptive transfer of NK cells is a viable option in managing leukemia.

Adoptive NK cell transfer can be done in the HSCT or non-HSCT setting, and at the same time, it can be either autologous or allogeneic.193,194 As a therapeutic strategy, autologous NK cell adoptive transfer is based on the extraction of the patient’s own NK cell from the peripheral blood, which is then expanded ex vivo and transduced back to the patient. This has its advantages in terms of convenience of source of NK cells, independence from immunosuppressants, and low likelihood of GvHD.195 To generate sufficient and high-quality NK cells, cytokines such as IL-2, IL-12, IL-15, and IL-18 stimulate NK cells to enhance their effector functions and proliferative capabilities. However, the increased proliferative capacity does not necessarily lead to a significant therapeutic outcome, and this is due to the inhibitory effect of the patient’s HLA ligands.

In some cases, the quality of NK cells may be below par because of prior heavy pretreatment of patients, giving rise to poor effector functions. While this may be so in AML, autologous adoptive NK cell transfer has shown efficacy in solid tumors and some hematological cancers.196–198 Various strategies are being developed to restore NK cell function. Wang et al.,199 in their study, noted that increased levels of TGF-β1 impaired bone marrow NK cells, and the use of TGF-β1 inhibitors like galunisertib or anti-TGF-β1 antibodies could restore NK cell effector functions. Furthermore, Lirilumab, an anti-KIR antibody that potentiates NK cells, has been shown to enhance therapeutic response as a combination therapy in vitro and in vivo. However, the EFFIKIR randomized, double-blind 3-arm placebo-controlled trial (NCT01687387) failed to improve leukaemia-free survival in elderly AML patients.200–202 These findings have caused a shift from autologous NK cells to allogeneic NK cell transfer by researchers.

For allogeneic NK cell transfer, NK cells obtained from healthy, HLA-matched, or haploidentical donors are prepared and expanded under standard conditions (Fig. 3). The NK cells are derived from different sources like autologous transfer, including peripheral blood NK cells, umbilical cord blood NK cells, NK cell lines, and stem cell-derived NK.203 In their study, Ruggeri et al.190 showed that allogeneic NK cell transfer in AML patients induced a significant EFS. An increase in donor chimerism was observed, while a decrease in chimerism and relapse was noted in one AML patient in another study.204 Different clinical studies of allogeneic NK cell transfer in the HSCT setting have shown tolerability and good efficacy.205–208 Several patients may not be eligible for HSCT, but this has not hindered the development of allogeneic NK transfer outside the HSCT setting. Miller et al.209 performed allogeneic NK cell transfer outside the HSCT setting, and 5 out of 19 achieved complete remission; this was significantly higher in those with KIR–ligand mismatched donors. Modifications to their approach have been replicated in other studies.210,211 These methods equally have their challenges, which include low clinical-grade activation, lack of in vivo persistence, and problems with ex vivo expansion. In all, adoptive NK cell transfer appears to be a sound therapeutic strategy for AML for induction remission and CR maintenance.

CAR-NK cell therapy

Following the success of CAR-T cell therapy in managing B-cell precursor acute lymphoblastic leukaemia (ALL) and B-cell lymphoma, cellular therapy has shown much optimism in managing other neoplasms, including AML. Despite such optimism, CAR-T cell therapy is yet to become a reality in the management of AML due to adverse events like cytokine release syndrome (CRS).212–214 Other obstacles encountered with CAR-T cells include inefficiencies of T cell isolation, modification and expansion, and high costs.215 There is much enthusiasm that CAR-NK cells can prove a better alternative to CAR-T cells due to their shorter lifespan, favorable toxicity profile, and lower manufacturing costs.216 Though it has some advantages, it has yet to be translated into a treatment option. Some challenges are still faced, including a loss of targeted antigen, hostile tumor microenvironment, and tumor heterogeneity. However, with progress made in NK cell engineering and target design, it is expected to prove efficacious in future trials. A CAR-NK cell product created from universal cord blood (UCB) NK cells by Liu and his colleagues was transduced with a retroviral vector that expressed genes that encoded anti-CD19 CAR, interleukin-15, and inducible caspase 9 as a safety switch. These were infused into 11 patients with B cell lymphoma and chronic lymphocytic leukaemia (CLL) in a phase 1/2 trial. Out of the 11 patients in the study, 7 had a CR, and a remission of the Richter’s transformation was reported in one, but with persistence of the CLL. These clinical responses were seen within 30 days.

The CAR-NK cells persisted in the patients for about 12 months, and there were no reported adverse events like cytokine release syndrome, neurotoxicity, or GvHD.217 This reflects some optimism in AML management. In preclinical studies, allogeneic CAR.CD123-NK cells induced significant anti-leukemic activity in vitro against CD123+ AML cell lines and CD123+ primary blasts and ex vivo in animal models.218 Another study using CD33/FLT3 CAR-NK cells showed antileukemic activity against primary AML blasts and LSC-enriched target cell populations and demonstrated improved survival in an MV4-11 xenograft AML mouse model.219 The C-type lectin-like molecule 1 (CLL-1), which is a widespread expression in AML blasts, has also been seen as an ideal target for CAR-NK cells. A phase 1 clinical trial recruits patients for CAR-NK cell targeting of CLL-1 in AML (NCT06027853).

CAR-NK cell therapy has also shown efficacy in human clinical trials. In a first-in-human phase 1 clinical trial, Huang et al.220 infused anti-CD33 CAR-NK cells into 10 R/R AML patients after preconditioning with fludarabine and cyclophosphamide. 60% of the patients had a complete response 28 days after the infusion of the CAR-NK cells, and only one patient developed grade 2 CRS, which was alleviated with dexamethasone.220 There were no reported incidences of neurotoxicity or any other adverse events. Some CAR-NK preclinical and clinical studies are underway across different cancers.221–223 Unfortunately, the phase 1 NKG2D CAR-NK cell therapy for R/R AML (NCT05247957) patients was prematurely terminated.224 CAR-NK cell therapy is a prospective option for managing AML (Fig. 4).

Schematic representation of adoptive NK cell transfer for AML patient.
Fig. 4  Schematic representation of adoptive NK cell transfer for AML patient.

For the allogeneic NK cell transfer, NK cells are isolated from a healthy HLA-matched or haploidentical donor. After T cell depletion and in vitro expansion, it is infused into the AML patient. For the autologous NK cell transfer, after NK cells are isolated from the peripheral blood of the AML patient, there shall be T cell depletion, followed by in vitro expansion before it is infused back into the patient. In CAR-NK cell therapy, NK cells can be harvested from different sources and engineered to express specific receptors that recognize ligands on the AML cells, leading to their destruction. AML, acute myeloid leukemia; CAR, chimeric antigen receptors; HLA, human leukocyte antigen; IL, interleukins; NK, natural killer.

Antibodies

The receptor-ligand interaction and antibody-dependent cellular cytotoxicity are two pivotal mechanisms for activating NK cells. Leveraging antibody mediation in these processes has emerged as a viable therapeutic strategy in AML. This can be achieved by targeting tumor-associated antigens or inhibiting NK cell receptors using specific antibodies.

Antibodies targeting tumor-associated antigens

The primary mechanism involves the induction of antibody-dependent cell-mediated cytotoxicity (ADCC) by NK cells. While unconjugated antibodies alone have shown limited efficacy, the engineering of antibodies to enhance their Fc regions can significantly improve their affinity for CD16, a receptor on NK cells that mediates ADCC. Preclinical studies have demonstrated the potential of these antibody-mediated actions. For example, Riegg et al.225 developed an anti-tumor antibody targeting CD133, a protein commonly found on the surface of B-ALL cells. This antibody specifically activated NK cells to lyse B-ALL cells. Similarly, Koerner et al.226 applied this molecule in studies involving AML cells and xenotransplanted mice, achieving cell lysis of CD133-expressing AML cells. Steinbacher et al.227 used Fc-optimized NKG2D-immunoglobulin G fusion proteins to activate NK cells against leukemia cell lines, including AML and primary AML cells, showing significant activity.

These studies underscore the effectiveness of using Fc-optimized antibodies against specific antigens expressed on AML cells as a promising therapeutic option. This approach enhances the innate immune response and offers a targeted method to combat leukemia cells by harnessing the natural cytotoxic functions of NK cells.

The antibody-drug conjugates and antibody-radio conjugate are promising therapeutic strategies for enhancing antibody potency. Gemtuzumab ozogamicin, a calicheamicin conjugate of anti-CD33 antibody, is approved to manage AML.228,229 Some other antibodies with conjugates such as CD13, FLT3, and CLL-1 and antibodies combined with NK cell transfer have shown promise for the management of AML.230–232

Antibodies targeting NK cell inhibitory receptors

NK cell’s inhibitory receptors are essential in immune cell recognition and tumor escape.233 These inhibitory receptors, including the MHC-I-specific inhibitory receptors (KIRs, LIRs) and immune checkpoints (PD-1, CTLA-4, TIGIT, Siglec-7, TIM-3), are known to cause NK cell dysfunction, as earlier discussed. While anti-KIRs have not proved to be efficacious in clinical trials, checkpoint inhibitors approved for some solid tumors have shown some efficacy in AML.201,202,234 In a phase 2 trial of nivolumab and azacitidine in pre-treated AML patients, the ORR was 33% with a CR of 22%.235 Another phase 2 trial of nivolumab in combination with cyclophosphamide in R/R AML patients has recently been concluded (NCT03417154). As for the anti-TIM-3 antibody sabatolimab (MBG453) in the phase 1 clinical study (with decitabine or azacitidine), it was safe and well tolerated in higher-risk myelodysplastic syndrome and AML.236,237 Of 11 clinical studies involving TIM-3 inhibitors in AML/MDS, three have completed recruitment (NCT03066648, NCT03946670, and NCT04266301).

BiKE and TriKE

BiKE and TriKE represent an innovative class of potential immunotherapeutic agents. These agents function as the NK cell counterparts to the bispecific T cell engager (BiTE), serving as immunological synapses between NK cells and cancer cells, similarly to how T cell engagers operate. T cell engagers activate T cells, leading to proliferation, cytokine release, and cancer cell death by bypassing the T cell receptor and MHC contact. While BiTE has been highly effective in managing hematological malignancies, it is associated with severe adverse events such as CRS and Immune effector cell-associated neurotoxicity syndrome (ICANS), which can cause significant morbidity and mortality.

Conversely, NK cell engagers primarily activate NK cells through cell surface receptors such as CD16, NKp46, or NKG2D.238–241 BiKEs and TriKEs have demonstrated promising activity against several cancers. Preclinical studies of a CD16xCD33 BiKE have shown that it can adequately activate NK cells, destroying AML cell lines and primary AML cells.242 A TriKE incorporating a modified human IL-15 into the CD16x CD33 BiKE has been shown to induce significant NK cell cytotoxicity, degranulation, and cytokine production against CD33+ HL-60 cells.241 Further studies on second-generation TriKEs have indicated that they are more potent than the first-generation and can induce cell death in patient-derived xenograft AML tumor models, as well as in both AML cell lines and primary patient-derived AML blasts.243–246

Additionally, Reusing et al.247 demonstrated that primary cells from pediatric AML and biphenotypic ALL responded positively to BiKE treatment. A phase 1/2 clinical trial (NCT03214666) using a designed TriKE reported significant reductions in bone marrow blast levels in patients with AML and MDS ) without the need for costly progenitor-derived or autologous/allogeneic cell therapies.248 These findings suggest that BiKEs and TriKEs, like their cousin BiTE, hold substantial potential in the management of AML, offering a targeted and effective approach to cancer immunotherapy.

Cytokines

In the developmental spectrum of NK cells, IL-2, IL-12, IL-15, IL-18, and IL-21 are critical players in the proliferation, activation, and effector functions of NK cells. IL-2 was the first cytokine shown to enhance NK cell activity, and to date, is the only Food and Drug Administration approved cytokine for the treatment of cancer patients. However, IL-15 is another very promising cytokine for activating NK cells. It is reported that in the ex vivo stimulation of NK cells in AML patients, 50 ng/mL of IL-15 or 10 ng/mL of IL-2 was optimal for the recovery of its function through the upregulation of activating receptors NKp30, NKp46, NKG2C, and NKG2D.249–251 Though it can expand and activate NK cells, studies have shown that 1L-2 may not have adequate clinical efficacy as a monotherapy in AML patients.252,253 However, IL-2, in conjunction with other therapies, has shown clinical efficacy in AML, especially as a maintenance therapy.254–257 Clinical studies have shown that IL-15 expands NK cells in cancer; however, high expression of IL-15 is reportedly linked with CNS disease and neurocognitive impairment in ALL.258–263 IL-15 has also been shown to increase the cytotoxicity of NK cells in patients with AML.264 In a phase 1 clinical trial, the IL-15 superagonist complex ALT-803 given as a monotherapy to AML patients who relapsed after allogeneic HSCT was observed to be safe and well-tolerated and with one CR (NCT01885897).265 A phase 1 trial of ALT-803 in solid tumors also produced a significant rise in NK cell numbers (NCT01727076).266 However, a recent clinical study by Berrien-Elliott et al.267 reported that systemic IL-15 can promote allogeneic cell rejection in R/R AML patients treated with natural killer cell adoptive therapy (NCT03050216 and NCT01898793). Recently, CAR-NK cells that co-expressed transgenes for the NKG2D CAR and IL-15 were developed, and it shows enhanced in vitro and in vivo activity in an AML mouse model.268 Regarding IL-21, a membrane-bound 1L-21 adoptive NK product was shown to reduce AML burden in vivo and had better OS in human subjects with AML.269 IL-21 was also found to inhibit primary AML stem cells in vitro with the enhancement of cytarabine treatment.270 Currently, two studies are recruiting for IL-21 trial in AML (NCT04220684) (NCT02809092).271,272

NK cells pre-activated with a cocktail of cytokines (IL-12, IL-15, and IL-18) have demonstrated sustained anti-leukemia responses to restimulation, maintaining effectiveness for weeks to months, regardless of inhibitory KIR-KIR ligand interactions. These cytokine-induced memory-like NK cells are reported to possess significant antineoplastic potential. A clinical trial involving the adoptive transfer of cytokine-induced memory-like (CIML) NK cells in R/R AML patients has shown that this approach can induce remission without serious adverse events.273 Further, an ongoing clinical trial involving donor transfer of CIML NK cells (NCT03068819) targeting R/R pediatric and young adult AML patients has provided encouraging data, reporting sustained CR.274,275 These findings highlight cytokine-induced NK cell products as promising therapeutic candidates for AML management. Details of these clinical trials are summarized in Table 2.

Table 2

Some clinical trials of NK cells cellular therapies

IdentifierPhaseConditionNK cell sourceInterventionStatusOutcome
NCT02809092I/IIR/R AMLHaploidentical NKBefore treatment with chemotherapyCompleted78.6% overall response; 50.0% CR; CNS responses in 4 patients
NCT01385423IRefractory AMLHaploidentical NKBefore treatment with lymphodepleting chemotherapy; After treatment with rhIL-15 intravenously (0.3–1.0 mg/kg)CompletedRobust NK expansion in 36% of patients at day 14; CR in 32% of patients
NCT00703820IIPaediatric AMLHaploidentical NKBefore treatment with lymphodepleting chemotherapy and rhIL-2 subcutaneouslyCompletedNone
NCT02763475IIPaediatric AMLHaploidentical NKBefore treatment with lymphodepleting chemotherapy and rhIL-2 subcutaneouslyCompletedCR in 6 of 7 patients
NCT05247957IR/R AMLCAR-NK cellPretreatedNot provided
NCT05272293I/IIPaediatric AMLHaploidentical NKPretreatedRecruitingNot provided
NCT05256277IR/R AML adultsCIML NK cellsPretreatedNot provided
NCT02727803IIAML, MDS, etcUCB-derived HSPC-NK cellTreated with Busulfan, Clofarabine, Cyclophosphamide, Fludarabine Phosphate, Melphalan, RituximabRecruitingNot provided
NCT01823198I/IIAML, MDS, etcPBMC-derived NK cellIL-2, Busulfan, FludarabineCompletedNot provided
NCT04221971IAML adultsPBMC-derived NK cellPretreatedCompleted1/3 with MRD negative, low dose group; 3/4 response with 1 case of extramedullary recurrence of AML turned negative, middle dose group.
NCT04310592IAML adultsPlacental-derived HSPC-NK cell (CYNK-001)PretreatedRecruitingNot provided
NCT04623944IAML adultsCar-NK cell (NKX101)PretreatedRecruitingNot provided
NCT04901416IAML adultsPBMC-derived NK cell (DVX201)PretreatedRecruitingNot provided
NCT04347616I/IIAMLUCB-NK cells + 1L-2PretreatedRecruitingNot provided
NCT05008575IR/R AMLCAR-NK cell (Anti-CD33)PretreatedRecruitingNot provided
NCT05215015IAMLCAR-NK cell (Anti-CD33/CLL1)PretreatedRecruitingNot provided
NCT04220684IAMLHaploidentical NK cell (IL-21 expanded)PretreatedRecruitingNot provided
NCT05333705IAMLPBMC/ UCB NK cellRecruitingNot provided
NCT04836390IPaediatric AMLHaploidentical NK cellEnrolling by invitationNot provided
NCT03821519I/IIAML, MDS etcCIML NK cellsPretreated (with allo-HSCT)RecruitingNot provided

Nanoparticles in enhancing NK cell therapy

New avenues are being explored for NK cell-based therapies. One such area is nanotechnology and nanomedicine. Nanotechnology is used to see its feasibility in NK cell expansion and activation. This can be done in several ways, including enhancing NK cell activity through nanoparticle-assisted immunomodulation, enhancing NK cell homing by nanoparticles, and activating NKG2D receptor by nanoparticles, etc.276,277 Several NK cell-based nano-immunotherapies for cancer are actively being developed, and one is currently in phase 2 trial.276 In a study on NK cells, Sanz-Ortega and her colleagues used magnetic nanoparticles to improve the targeting of adoptively transferred NK cells without altering their function.278 Selenium-containing nanoparticles were used in a study to enhance NK cell function.279 Nanoengagers were shown to be more effective in activating NK cells than antibodies. In addition, they could augment both NK-activating agents and chemotherapy to achieve a greater intensity of chemoimmunotherapy.280 Nanoengagers were also created for T cells against an AML xenograft model, which effectively activated T cells and induced AML cell death in vitro and in vivo.281 This shows the potential of nano-immunotherapies in the management of hematological malignancies. Very recently, Zeinabad engineered an NK cell mimic nanoparticle, which was functionalized against an anti-CD38 antibody (Daratumumab). It showed in vitro activity against AML cell lines, patient-derived AML cells ex vivo, and CD38-positive AML cells in vivo in a disseminated AML xenograft model.282 This same nanocoupling was also successfully used to target some hematological cancer cell lines.283 NK cell-based nano-immunotherapy is still in its infancy, but it is believed it can be one of the arsenals against AML shortly.

Conclusions and future perspective

NK cell-based therapies have shown potential as viable and strategic therapeutics in managing AML in the future. So far, the various preclinical and clinical studies on NK cells show a challenging but achievable feat. However, it is a priority to get the different NK cell therapeutic forms to do what they are for against a highly heterogeneous enemy like AML. Compared to T cell therapies, NK cells have some advantages. NK cell tumor detection is not strictly based on MHC recognition but can mediate ADCC. NK cells also provide a better safety profile than T cell therapies, including a lower incidence of GvHD, CRS, and ICANS. Though NK cells have a limited lifespan, they are easy to prepare under good manufacturing practice standards, implying an “off the shelf” benefit and a universal administration for managing patients in a short period. However, NK cell cellular-based therapies are still faced with some challenges, including ensuring sustained in vivo expansion and proliferation of NK cells due to their short lifespan in patients, which leads to a short response duration. How can the various immune escape mechanisms used by AML be stopped to evade detection and cell death, especially through the creation of an immunosuppressive tumor microenvironment?

The tumor microenvironment in AML is a complex arena that impairs NK cell function. For example, myeloid-derived suppressive cells, which are found within the tumor microenvironment, can produce immunosuppressive factors such as IL-10, TGF-β, and IL-4, which are capable of inhibiting the expression of NKp30 and NKG2D that are important in NK cell tumor cells recognition and destruction. AML cells also alter glucose utilization, enabling them to survive hypoxic conditions. Glucose is vital for NK cell metabolism and is thus reduced, leading to NK cell dysfunction. Increasing lactic acid production in the microenvironment is a potent inhibitor of NK cell effector function and viability. Thus, this microenvironment hostility has been shown to affect NK cell therapy, especially CAR-NK cells. TGF-β, which plays an inhibitory role in NK cell tumor cell recognition, can be neutralized by engineering CAR-NK cells that lack TGF-β receptor expression. Moreover, catalase can attenuate hypoxia in the microenvironment, reducing the effect of lactic acid and hydrogen peroxide on NK cells, ultimately improving NK cell therapy. In addition, cytokines such as IL-15 and IL-21 can enhance NK cell cytotoxicity in tumor sites, while IL-18 primed NK cells can also engage effector T cells through the help of DCs.

The efficient transduction of CAR-NK cells remains a critical issue that requires further exploration. While the field has seen several phase 1/2 clinical trials, initiating a phase 3 trial is imperative. This next step involves a well-designed, randomized clinical trial with an adequate sample size to determine the optimal dosing and therapeutic efficacy of each NK cell therapy in AML). Such a trial could also elucidate the most efficacious NK cell therapy for AML and address the timing of these therapies—whether AML patients should receive them during induction remission, consolidation, or as part of a maintenance regime, and how many cycles should be administered at each stage.

Considering our current understanding of various NK cellular therapies, there is potential for using them in combination therapies. Such combinations might enhance both the proliferative and effector functions of NK cells and their in vivo sustainability to effectively target AML cells. In particular, CIML NK cells could be valuable due to their ability to prolong the duration of NK cells in vivo. Furthermore, combining standard AML therapies with NK cellular therapies could provide synergistic effects that enhance the ability of NK cells to combat AML.

Immunomodulatory drugs, such as lenalidomide and thalidomide, have shown promise in enhancing NK cell functions. They achieve this by stimulating the release of IL-2 and IFN-γ from T cells and dendritic cells in the surrounding environment. Additionally, proteasome inhibitors like bortezomib can increase the sensitivity of AML cells to NK cell-mediated lysis, potentially improving clinical outcomes. Such strategic integration of therapies could lead to more effective, sustainable treatments for AML, capitalizing on the innate strengths of NK cells in cancer immunotherapy.

In conclusion, the treatment landscape for managing AML has expanded with the potential integration of NK cell cellular-based therapies. These therapies stand out among other cellular treatments due to their off-the-shelf availability, cost-effectiveness, and capability to recognize cancer cells without the constraint of MHC mechanisms. This attribute facilitates broader accessibility and potentially fewer adverse effects for various patients. The efficacy and safety of these therapies are highlighted in the clinical trial (NCT03056339) reported by Marin et al.,284 where CAR-NK cells were used to treat CD19+ B cell malignancies. In this trial, no notable adverse events such as ICANS, CRS, or GVHD were observed. This evidence further solidifies the favorable safety profile of NK cell-based cellular therapies, promising a valuable addition to the arsenal against AML.

NK cell cellular-based therapies have a bright prospect in managing AML, and with more clinical research, it may soon be a reality.

Declarations

Acknowledgement

None.

Funding

The author did not receive any funding for this work.

Conflict of interest

The author declares that he has no competing interests to declare.

Authors’ contributions

OOI is the sole author of the manuscript.

References

  1. Levis M. Midostaurin approved for FLT3-mutated AML. Blood 2017;129(26):3403-3406 View Article PubMed/NCBI
  2. Lai C, Doucette K, Norsworthy K. Recent drug approvals for acute myeloid leukemia. J Hematol Oncol 2019;12(1):100 View Article PubMed/NCBI
  3. National Cancer Institute. Cancer Stat Facts: Leukemia - Acute Myeloid Leukemia (AML). Available from: https://seer.cancer.gov/statfacts/html/amyl.html. Accessed November 15, 2022
  4. Vago L, Gojo I. Immune escape and immunotherapy of acute myeloid leukemia. J Clin Invest 2020;130(4):1552-1564 View Article PubMed/NCBI
  5. Isidori A, Cerchione C, Daver N, DiNardo C, Garcia-Manero G, Konopleva M, et al. Immunotherapy in Acute Myeloid Leukemia: Where We Stand. Front Oncol 2021;11:656218 View Article PubMed/NCBI
  6. Sambi M, Bagheri L, Szewczuk MR. Current Challenges in Cancer Immunotherapy: Multimodal Approaches to Improve Efficacy and Patient Response Rates. J Oncol 2019;2019:4508794 View Article PubMed/NCBI
  7. Kim SK, Cho SW. The Evasion Mechanisms of Cancer Immunity and Drug Intervention in the Tumor Microenvironment. Front Pharmacol 2022;13:868695 View Article PubMed/NCBI
  8. Ge Z, Wu S, Zhang Z, Ding S. Mechanism of tumor cells escaping from immune surveillance of NK cells. Immunopharmacol Immunotoxicol 2020;42(3):187-198 View Article PubMed/NCBI
  9. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 2020;20(11):651-668 View Article PubMed/NCBI
  10. Tang L, Huang Z, Mei H, Hu Y. Immunotherapy in hematologic malignancies: achievements, challenges and future prospects. Signal Transduct Target Ther 2023;8(1):306 View Article PubMed/NCBI
  11. Liu E, Tong Y, Dotti G, Shaim H, Savoldo B, Mukherjee M, et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 2018;32(2):520-531 View Article PubMed/NCBI
  12. Gang M, Marin ND, Wong P, Neal CC, Marsala L, Foster M, et al. CAR-modified memory-like NK cells exhibit potent responses to NK-resistant lymphomas. Blood 2020;136(20):2308-2318 View Article PubMed/NCBI
  13. Nguyen S, Lacan C, Roos-Weil D. [Allogeneic CAR-NK cells: A promising alternative to autologous CAR-T cells - State of the art, sources of NK cells, limits and perspectives]. Bull Cancer 2021;108(10S):S81-S91 View Article PubMed/NCBI
  14. Sabbah M, Jondreville L, Lacan C, Norol F, Vieillard V, Roos-Weil D, et al. CAR-NK Cells: A Chimeric Hope or a Promising Therapy?. Cancers (Basel) 2022;14(15):3839 View Article PubMed/NCBI
  15. Equipping NK Cells with CARs. Cancer Discov 2017;7(10):OF2 View Article PubMed/NCBI
  16. Davis ZB, Felices M, Verneris MR, Miller JS. Natural Killer Cell Adoptive Transfer Therapy: Exploiting the First Line of Defense Against Cancer. Cancer J 2015;21(6):486-491 View Article PubMed/NCBI
  17. Laskowski TJ, Biederstädt A, Rezvani K. Natural killer cells in antitumour adoptive cell immunotherapy. Nat Rev Cancer 2022;22(10):557-575 View Article PubMed/NCBI
  18. Tanaka J, Tanaka N, Wang YH, Mitsuhashi K, Ryuzaki M, Iizuka Y, et al. Phase I study of cellular therapy using ex vivo expanded natural killer cells from autologous peripheral blood mononuclear cells combined with rituximab-containing chemotherapy for relapsed CD20-positive malignant lymphoma patients. Haematologica 2020;105(4):e190-e193 View Article PubMed/NCBI
  19. Singh R, Gupta U, Srivastava P, Paladhi A, Sk UH, Hira SK, et al. γc cytokine-aided crosstalk between dendritic cells and natural killer cells together with doxorubicin induces a healer response in experimental lymphoma by downregulating FOXP3 and programmed cell death protein 1. Cytotherapy 2022;24(12):1232-1244 View Article PubMed/NCBI
  20. Gupta U, Hira SK, Singh R, Paladhi A, Srivastava P, Pratim Manna P. Essential role of TNF-α in gamma c cytokine aided crosstalk between dendritic cells and natural killer cells in experimental murine lymphoma. Int Immunopharmacol 2020;78:106031 View Article PubMed/NCBI
  21. Berjis A, Muthumani D, Aguilar OA, Pomp O, Johnson O, Finck AV, et al. Pretreatment with IL-15 and IL-18 rescues natural killer cells from granzyme B-mediated apoptosis after cryopreservation. Nat Commun 2024;15(1):3937 View Article PubMed/NCBI
  22. Pinto S, Pahl J, Schottelius A, Carter PJ, Koch J. Reimagining antibody-dependent cellular cytotoxicity in cancer: the potential of natural killer cell engagers. Trends Immunol 2022;43(11):932-946 View Article PubMed/NCBI
  23. Nikkhoi SK, Li G, Eleya S, Yang G, Vandavasi VG, Hatefi A. Bispecific killer cell engager with high affinity and specificity toward CD16a on NK cells for cancer immunotherapy. Front Immunol 2022;13:1039969 View Article PubMed/NCBI
  24. Le Roy A, Prébet T, Castellano R, Goubard A, Riccardi F, Fauriat C, et al. Immunomodulatory Drugs Exert Anti-Leukemia Effects in Acute Myeloid Leukemia by Direct and Immunostimulatory Activities. Front Immunol 2018;9:977 View Article PubMed/NCBI
  25. Felices M, Lenvik TR, Davis ZB, Miller JS, Vallera DA. Generation of BiKEs and TriKEs to Improve NK Cell-Mediated Targeting of Tumor Cells. Methods Mol Biol 2016;1441:333-346 View Article PubMed/NCBI
  26. Luna JI, Grossenbacher SK, Sturgill IR, Ames E, Judge SJ, Bouzid LA, et al. Bortezomib Augments Natural Killer Cell Targeting of Stem-Like Tumor Cells. Cancers (Basel) 2019;11(1):85 View Article PubMed/NCBI
  27. Allison M, Mathews J, Gilliland T, Mathew SO. Natural Killer Cell-Mediated Immunotherapy for Leukemia. Cancers (Basel) 2022;14(3):843 View Article PubMed/NCBI
  28. Collins SM, Bakan CE, Swartzel GD, Hofmeister CC, Efebera YA, Kwon H, et al. Elotuzumab directly enhances NK cell cytotoxicity against myeloma via CS1 ligation: evidence for augmented NK cell function complementing ADCC. Cancer Immunol Immunother 2013;62(12):1841-1849 View Article PubMed/NCBI
  29. Langers I, Renoux VM, Thiry M, Delvenne P, Jacobs N. Natural killer cells: role in local tumor growth and metastasis. Biologics 2012;6:73-82 View Article PubMed/NCBI
  30. Della Chiesa M, Pesce S, Muccio L, Carlomagno S, Sivori S, Moretta A, et al. Features of Memory-Like and PD-1(+) Human NK Cell Subsets. Front Immunol 2016;7:351 View Article PubMed/NCBI
  31. Cooper MA, Fehniger TA, Turner SC, Chen KS, Ghaheri BA, Ghayur T, et al. Human natural killer cells: a unique innate immunoregulatory role for the CD56(bright) subset. Blood 2001;97(10):3146-3151 View Article PubMed/NCBI
  32. Cooper MA, Fehniger TA, Caligiuri MA. The biology of human natural killer-cell subsets. Trends Immunol 2001;22(11):633-640 View Article PubMed/NCBI
  33. Duggan MC, Campbell AR, McMichael EL, Opheim KS, Levine KM, Bhave N, et al. Co-stimulation of the fc receptor and interleukin-12 receptor on human natural killer cells leads to increased expression of cd25. Oncoimmunology 2018;7(2):e1381813 View Article PubMed/NCBI
  34. Wagner JA, Rosario M, Romee R, Berrien-Elliott MM, Schneider SE, Leong JW, et al. CD56bright NK cells exhibit potent antitumor responses following IL-15 priming. J Clin Invest 2017;127(11):4042-4058 View Article PubMed/NCBI
  35. Rubio MT, Dhuyser A, Nguyen S. Role and Modulation of NK Cells in Multiple Myeloma. Hemato 2021;2(2):167-181 View Article
  36. Passlick B, Izbicki JR, Waydhas C, Nast-Kolb D, Schweiberer L, Ziegler-Heitbrock HW. Posttraumatic splenectomy does not influence human peripheral blood mononuclear cell subsets. J Clin Lab Immunol 1991;34(4):157-161 PubMed/NCBI
  37. Ramos SB, Garcia AB, Viana SR, Voltarelli JC, Falcão RP. Phenotypic and functional evaluation of natural killer cells in thymectomized children. Clin Immunol Immunopathol 1996;81(3):277-281 View Article PubMed/NCBI
  38. Cavalcanti NV, Palmeira P, Jatene MB, de Barros Dorna M, Carneiro-Sampaio M. Early Thymectomy Is Associated With Long-Term Impairment of the Immune System: A Systematic Review. Front Immunol 2021;12:774780 View Article PubMed/NCBI
  39. Colucci F, Caligiuri MA, Di Santo JP. What does it take to make a natural killer?. Nat Rev Immunol 2003;3(5):413-425 View Article PubMed/NCBI
  40. Moroso V, Famili F, Papazian N, Cupedo T, van der Laan LJ, Kazemier G, et al. NK cells can generate from precursors in the adult human liver. Eur J Immunol 2011;41(11):3340-3350 View Article PubMed/NCBI
  41. Freud AG, Becknell B, Roychowdhury S, Mao HC, Ferketich AK, Nuovo GJ, et al. A human CD34(+) subset resides in lymph nodes and differentiates into CD56bright natural killer cells. Immunity 2005;22(3):295-304 View Article PubMed/NCBI
  42. Ferlazzo G, Thomas D, Lin SL, Goodman K, Morandi B, Muller WA, et al. The abundant NK cells in human secondary lymphoid tissues require activation to express killer cell Ig-like receptors and become cytolytic. J Immunol 2004;172(3):1455-1462 View Article PubMed/NCBI
  43. Görgens A, Ludwig AK, Möllmann M, Krawczyk A, Dürig J, Hanenberg H, et al. Multipotent hematopoietic progenitors divide asymmetrically to create progenitors of the lymphomyeloid and erythromyeloid lineages. Stem Cell Reports 2014;3(6):1058-1072 View Article PubMed/NCBI
  44. Kohn LA, Hao QL, Sasidharan R, Parekh C, Ge S, Zhu Y, et al. Lymphoid priming in human bone marrow begins before expression of CD10 with upregulation of L-selectin. Nat Immunol 2012;13(10):963-971 View Article PubMed/NCBI
  45. Adolfsson J, Månsson R, Buza-Vidas N, Hultquist A, Liuba K, Jensen CT, et al. Identification of Flt3+ lympho-myeloid stem cells lacking erythro-megakaryocytic potential a revised road map for adult blood lineage commitment. Cell 2005;121(2):295-306 View Article PubMed/NCBI
  46. Kondo M. Lymphoid and myeloid lineage commitment in multipotent hematopoietic progenitors. Immunol Rev 2010;238(1):37-46 View Article PubMed/NCBI
  47. Karsunky H, Inlay MA, Serwold T, Bhattacharya D, Weissman IL. Flk2+ common lymphoid progenitors possess equivalent differentiation potential for the B and T lineages. Blood 2008;111(12):5562-5570 View Article PubMed/NCBI
  48. Inlay MA, Bhattacharya D, Sahoo D, Serwold T, Seita J, Karsunky H, et al. Ly6d marks the earliest stage of B-cell specification and identifies the branchpoint between B-cell and T-cell development. Genes Dev 2009;23(20):2376-2381 View Article PubMed/NCBI
  49. Stokic-Trtica V, Diefenbach A, Klose CSN. NK Cell Development in Times of Innate Lymphoid Cell Diversity. Front Immunol 2020;11:813 View Article PubMed/NCBI
  50. Lopes N, Vivier E, Narni-Mancinelli E. Natural killer cells and type 1 innate lymphoid cells in cancer. Semin Immunol 2023;66:101709 View Article PubMed/NCBI
  51. Vivier E, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, et al. Innate Lymphoid Cells: 10 Years On. Cell 2018;174(5):1054-1066 View Article PubMed/NCBI
  52. Chiossone L, Dumas PY, Vienne M, Vivier E. Natural killer cells and other innate lymphoid cells in cancer. Nat Rev Immunol 2018;18(11):671-688 View Article PubMed/NCBI
  53. Yang Q, Li F, Harly C, Xing S, Ye L, Xia X, et al. TCF-1 upregulation identifies early innate lymphoid progenitors in the bone marrow. Nat Immunol 2015;16(10):1044-1050 View Article PubMed/NCBI
  54. Harly C, Kenney D, Ren G, Lai B, Raabe T, Yang Q, et al. The transcription factor TCF-1 enforces commitment to the innate lymphoid cell lineage. Nat Immunol 2019;20(9):1150-1160 View Article PubMed/NCBI
  55. Klose CSN, Flach M, Möhle L, Rogell L, Hoyler T, Ebert K, et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell 2014;157(2):340-356 View Article PubMed/NCBI
  56. Yagi R, Zhong C, Northrup DL, Yu F, Bouladoux N, Spencer S, et al. The transcription factor GATA3 is critical for the development of all IL-7Rα-expressing innate lymphoid cells. Immunity 2014;40(3):378-388 View Article PubMed/NCBI
  57. Gordon SM, Chaix J, Rupp LJ, Wu J, Madera S, Sun JC, et al. The transcription factors T-bet and Eomes control key checkpoints of natural killer cell maturation. Immunity 2012;36(1):55-67 View Article PubMed/NCBI
  58. Kiekens L, Van Loocke W, Taveirne S, Wahlen S, Persyn E, Van Ammel E, et al. T-BET and EOMES Accelerate and Enhance Functional Differentiation of Human Natural Killer Cells. Front Immunol 2021;12:732511 View Article PubMed/NCBI
  59. Schroeder JH, Howard JK, Lord GM. Transcription factor-driven regulation of ILC1 and ILC3. Trends Immunol 2022;43(7):564-579 View Article PubMed/NCBI
  60. Ebihara T, Taniuchi I. Transcription Factors in the Development and Function of Group 2 Innate Lymphoid Cells. Int J Mol Sci 2019;20(6):1377 View Article PubMed/NCBI
  61. Korchagina AA, Shein SA, Koroleva E, Tumanov AV. Transcriptional control of ILC identity. Front Immunol 2023;14:1146077 View Article PubMed/NCBI
  62. Zhong C, Cui K, Wilhelm C, Hu G, Mao K, Belkaid Y, et al. Group 3 innate lymphoid cells continuously require the transcription factor GATA-3 after commitment. Nat Immunol 2016;17(2):169-178 View Article PubMed/NCBI
  63. Bi J, Wang X. Molecular Regulation of NK Cell Maturation. Front Immunol 2020;11:1945 View Article PubMed/NCBI
  64. Yu J, Freud AG, Caligiuri MA. Location and cellular stages of natural killer cell development. Trends Immunol 2013;34(12):573-582 View Article PubMed/NCBI
  65. Becknell B, Caligiuri MA. Interleukin-2, interleukin-15, and their roles in human natural killer cells. Adv Immunol 2005;86:209-239 View Article PubMed/NCBI
  66. Scoville SD, Mundy-Bosse BL, Zhang MH, Chen L, Zhang X, Keller KA, et al. A Progenitor Cell Expressing Transcription Factor RORγt Generates All Human Innate Lymphoid Cell Subsets. Immunity 2016;44(5):1140-1150 View Article PubMed/NCBI
  67. Wang D, Malarkannan S. Transcriptional Regulation of Natural Killer Cell Development and Functions. Cancers (Basel) 2020;12(6):1591 View Article PubMed/NCBI
  68. Di Vito C, Mikulak J, Mavilio D. On the Way to Become a Natural Killer Cell. Front Immunol 2019;10:1812 View Article PubMed/NCBI
  69. Poli A, Michel T, Thérésine M, Andrès E, Hentges F, Zimmer J. CD56bright natural killer (NK) cells: an important NK cell subset. Immunology 2009;126(4):458-465 View Article PubMed/NCBI
  70. Kared H, Martelli S, Tan SW, Simoni Y, Chong ML, Yap SH, et al. Adaptive NKG2C(+)CD57(+) Natural Killer Cell and Tim-3 Expression During Viral Infections. Front Immunol 2018;9:686 View Article PubMed/NCBI
  71. Pesce S, Squillario M, Greppi M, Loiacono F, Moretta L, Moretta A, et al. New miRNA Signature Heralds Human NK Cell Subsets at Different Maturation Steps: Involvement of miR-146a-5p in the Regulation of KIR Expression. Front Immunol 2018;9:2360 View Article PubMed/NCBI
  72. Wagner JA, Fehniger TA. Human Adaptive Natural Killer Cells: Beyond NKG2C. Trends Immunol 2016;37(6):351-353 View Article PubMed/NCBI
  73. Elliott JM, Yokoyama WM. Unifying concepts of MHC-dependent natural killer cell education. Trends Immunol 2011;32(8):364-372 View Article PubMed/NCBI
  74. Kim S, Poursine-Laurent J, Truscott SM, Lybarger L, Song YJ, Yang L, et al. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature 2005;436(7051):709-713 View Article PubMed/NCBI
  75. Yokoyama WM, Kim S. Licensing of natural killer cells by self-major histocompatibility complex class I. Immunol Rev 2006;214:143-154 View Article PubMed/NCBI
  76. Raulet DH, Vance RE. Self-tolerance of natural killer cells. Nat Rev Immunol 2006;6(7):520-531 View Article PubMed/NCBI
  77. He Y, Tian Z. NK cell education via nonclassical MHC and non-MHC ligands. Cell Mol Immunol 2017;14(4):321-330 View Article PubMed/NCBI
  78. Joncker NT, Raulet DH. Regulation of NK cell responsiveness to achieve self-tolerance and maximal responses to diseased target cells. Immunol Rev 2008;224:85-97 View Article PubMed/NCBI
  79. Brodin P, Kärre K, Höglund P. NK cell education: not an on-off switch but a tunable rheostat. Trends Immunol 2009;30(4):143-149 View Article PubMed/NCBI
  80. Shifrin N, Raulet DH, Ardolino M. NK cell self tolerance, responsiveness and missing self recognition. Semin Immunol 2014;26(2):138-144 View Article PubMed/NCBI
  81. Martin MP, Single RM, Wilson MJ, Trowsdale J, Carrington M. KIR haplotypes defined by segregation analysis in 59 Centre d’Etude Polymorphisme Humain (CEPH) families. Immunogenetics 2008;60(12):767-774 View Article PubMed/NCBI
  82. Pende D, Falco M, Vitale M, Cantoni C, Vitale C, Munari E, et al. Killer Ig-Like Receptors (KIRs): Their Role in NK Cell Modulation and Developments Leading to Their Clinical Exploitation. Front Immunol 2019;10:1179 View Article PubMed/NCBI
  83. Sivori S, Parolini S, Falco M, Marcenaro E, Biassoni R, Bottino C, et al. 2B4 functions as a co-receptor in human NK cell activation. Eur J Immunol 2000;30(3):787-793 View Article PubMed/NCBI
  84. Bottino C, Falco M, Parolini S, Marcenaro E, Augugliaro R, Sivori S, et al. NTB-A [correction of GNTB-A], a novel SH2D1A-associated surface molecule contributing to the inability of natural killer cells to kill Epstein-Barr virus-infected B cells in X-linked lymphoproliferative disease. J Exp Med 2001;194(3):235-246 View Article PubMed/NCBI
  85. Watzl C, Claus M. WhatSAP - 2B4 sends mixed messages in the absence of SAP. Eur J Immunol 2014;44(5):1281-1284 View Article PubMed/NCBI
  86. Meazza R, Tuberosa C, Cetica V, Falco M, Loiacono F, Parolini S, et al. XLP1 inhibitory effect by 2B4 does not affect DNAM-1 and NKG2D activating pathways in NK cells. Eur J Immunol 2014;44(5):1526-1534 View Article PubMed/NCBI
  87. Marcenaro E, Augugliaro R, Falco M, Castriconi R, Parolini S, Sivori S, et al. CD59 is physically and functionally associated with natural cytotoxicity receptors and activates human NK cell-mediated cytotoxicity. Eur J Immunol 2003;33(12):3367-3376 View Article PubMed/NCBI
  88. Li L, Yu S, Liu S, Meng F, Ren X, Liu Z, et al. The expression and clinical significance of CD59 and FLAER in Chinese adult AML patients. J Clin Lab Anal 2022;36(1):e24145 View Article PubMed/NCBI
  89. Pende D, Spaggiari GM, Marcenaro S, Martini S, Rivera P, Capobianco A, et al. Analysis of the receptor-ligand interactions in the natural killer-mediated lysis of freshly isolated myeloid or lymphoblastic leukemias: evidence for the involvement of the Poliovirus receptor (CD155) and Nectin-2 (CD112). Blood 2005;105(5):2066-2073 View Article PubMed/NCBI
  90. Li J, Whelan S, Kotturi MF, Meyran D, D’Souza C, Hansen K, et al. PVRIG is a novel natural killer cell immune checkpoint receptor in acute myeloid leukemia. Haematologica 2021;106(12):3115-3124 View Article PubMed/NCBI
  91. Li Y, Zhang Y, Cao G, Zheng X, Sun C, Wei H, et al. Blockade of checkpoint receptor PVRIG unleashes anti-tumor immunity of NK cells in murine and human solid tumors. J Hematol Oncol 2021;14(1):100 View Article PubMed/NCBI
  92. Sanchez-Correa B, Valhondo I, Hassouneh F, Lopez-Sejas N, Pera A, Bergua JM, et al. DNAM-1 and the TIGIT/PVRIG/TACTILE Axis: Novel Immune Checkpoints for Natural Killer Cell-Based Cancer Immunotherapy. Cancers (Basel) 2019;11(6):877 View Article PubMed/NCBI
  93. Bolm L, Petruch N, Sivakumar S, Annels NE, Frampton AE. Gene of the month: T-cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT). J Clin Pathol 2022;75(4):217-221 View Article PubMed/NCBI
  94. Liu G, Zhang Q, Yang J, Li X, Xian L, Li W, et al. Increased TIGIT expressing NK cells with dysfunctional phenotype in AML patients correlated with poor prognosis. Cancer Immunol Immunother 2022;71(2):277-287 View Article PubMed/NCBI
  95. Kaito Y, Sugimoto E, Nakamura F, Tsukune Y, Sasaki M, Yui S, et al. Immune checkpoint molecule DNAM-1/CD112 axis is a novel target for natural killer-cell therapy in acute myeloid leukemia. Haematologica 2024;109(4):1107-1120 View Article PubMed/NCBI
  96. Dai YJ, He SY, Hu F, Li XP, Zhang JM, Chen SL, et al. Bone marrow infiltrated natural killer cells predicted the anti-leukemia activity of MCL1 or BCL2 inhibitors in acute myeloid leukemia. Mol Cancer 2021;20(1):8 View Article PubMed/NCBI
  97. Gwalani LA, Orange JS. Single Degranulations in NK Cells Can Mediate Target Cell Killing. J Immunol 2018;200(9):3231-3243 View Article PubMed/NCBI
  98. Kabanova A, Zurli V, Baldari CT. Signals Controlling Lytic Granule Polarization at the Cytotoxic Immune Synapse. Front Immunol 2018;9:307 View Article PubMed/NCBI
  99. Hsu HT, Carisey AF, Orange JS. Measurement of Lytic Granule Convergence After Formation of an NK Cell Immunological Synapse. Methods Mol Biol 2017;1584:497-515 View Article PubMed/NCBI
  100. Chen X, Trivedi PP, Ge B, Krzewski K, Strominger JL. Many NK cell receptors activate ERK2 and JNK1 to trigger microtubule organizing center and granule polarization and cytotoxicity. Proc Natl Acad Sci U S A 2007;104(15):6329-6334 View Article PubMed/NCBI
  101. Mace EM, Dongre P, Hsu HT, Sinha P, James AM, Mann SS, et al. Cell biological steps and checkpoints in accessing NK cell cytotoxicity. Immunol Cell Biol 2014;92(3):245-255 View Article PubMed/NCBI
  102. Stinchcombe JC, Majorovits E, Bossi G, Fuller S, Griffiths GM. Centrosome polarization delivers secretory granules to the immunological synapse. Nature 2006;443(7110):462-465 View Article PubMed/NCBI
  103. Osińska I, Popko K, Demkow U. Perforin: an important player in immune response. Cent Eur J Immunol 2014;39(1):109-115 View Article PubMed/NCBI
  104. Barry M, Heibein JA, Pinkoski MJ, Lee SF, Moyer RW, Green DR, et al. Granzyme B short-circuits the need for caspase 8 activity during granule-mediated cytotoxic T-lymphocyte killing by directly cleaving Bid. Mol Cell Biol 2000;20(11):3781-3794 View Article PubMed/NCBI
  105. Wang H, Huang Y, He J, Zhong L, Zhao Y. Dual roles of granzyme B. J Immun 2021;94(3):e13086 View Article
  106. Guicciardi ME, Gores GJ. Life and death by death receptors. FASEB J 2009;23(6):1625-1637 View Article PubMed/NCBI
  107. Bao Q, Shi Y. Apoptosome: a platform for the activation of initiator caspases. Cell Death Differ 2007;14(1):56-65 View Article PubMed/NCBI
  108. Bui VT, Tseng HC, Kozlowska A, Maung PO, Kaur K, Topchyan P, et al. Augmented IFN-γ and TNF-α Induced by Probiotic Bacteria in NK Cells Mediate Differentiation of Stem-Like Tumors Leading to Inhibition of Tumor Growth and Reduction in Inflammatory Cytokine Release; Regulation by IL-10. Front Immunol 2015;6:576 View Article PubMed/NCBI
  109. Jewett A, Kos J, Kaur K, Safaei T, Sutanto C, Chen W, et al. Natural Killer Cells: Diverse Functions in Tumor Immunity and Defects in Pre-neoplastic and Neoplastic Stages of Tumorigenesis. Mol Ther Oncolytics 2020;16:41-52 View Article PubMed/NCBI
  110. Walzer T, Dalod M, Robbins SH, Zitvogel L, Vivier E. Natural-killer cells and dendritic cells: “l’union fait la force”. Blood 2005;106(7):2252-2258 View Article PubMed/NCBI
  111. Tabellini G, Patrizi O, Dobbs K, Lougaris V, Baronio M, Coltrini D, et al. From Natural Killer Cell Receptor Discovery to Characterization of Natural Killer Cell Defects in Primary Immunodeficiencies. Front Immunol 2019;10:1757 View Article PubMed/NCBI
  112. Lougaris V, Patrizi O, Baronio M, Tabellini G, Tampella G, Damiati E, et al. NFKB1 regulates human NK cell maturation and effector functions. Clin Immunol 2017;175:99-108 View Article PubMed/NCBI
  113. Atsaves V, Leventaki V, Rassidakis GZ, Claret FX. AP-1 Transcription Factors as Regulators of Immune Responses in Cancer. Cancers (Basel) 2019;11(7):1037 View Article PubMed/NCBI
  114. Vaeth M, Feske S. NFAT control of immune function: New Frontiers for an Abiding Trooper. F1000Res 2018;7:260 View Article PubMed/NCBI
  115. Allen F, Bobanga ID, Rauhe P, Barkauskas D, Teich N, Tong C, et al. CCL3 augments tumor rejection and enhances CD8(+) T cell infiltration through NK and CD103(+) dendritic cell recruitment via IFNγ. Oncoimmunology 2018;7(3):e1393598 View Article PubMed/NCBI
  116. Morel PA, Ernst LK, Metes D. Functional CD32 molecules on human NK cells. Leuk Lymphoma 1999;35(1-2):47-56 View Article PubMed/NCBI
  117. Lanier LL, Ruitenberg JJ, Phillips JH. Functional and biochemical analysis of CD16 antigen on natural killer cells and granulocytes. J Immunol 1988;141(10):3478-3485 PubMed/NCBI
  118. Smyth MJ, Cretney E, Kelly JM, Westwood JA, Street SE, Yagita H, et al. Activation of NK cell cytotoxicity. Mol Immunol 2005;42(4):501-510 View Article PubMed/NCBI
  119. Lo Nigro C, Macagno M, Sangiolo D, Bertolaccini L, Aglietta M, Merlano MC. NK-mediated antibody-dependent cell-mediated cytotoxicity in solid tumors: biological evidence and clinical perspectives. Ann Transl Med 2019;7(5):105 View Article PubMed/NCBI
  120. Sun JC, Beilke JN, Lanier LL. Adaptive immune features of natural killer cells. Nature 2009;457(7229):557-561 View Article PubMed/NCBI
  121. Schlub TE, Sun JC, Walton SM, Robbins SH, Pinto AK, Munks MW, et al. Comparing the kinetics of NK cells, CD4, and CD8 T cells in murine cytomegalovirus infection. J Immunol 2011;187(3):1385-1392 View Article PubMed/NCBI
  122. O’Leary JG, Goodarzi M, Drayton DL, von Andrian UH. T cell- and B cell-independent adaptive immunity mediated by natural killer cells. Nat Immunol 2006;7(5):507-516 View Article PubMed/NCBI
  123. Foley B, Cooley S, Verneris MR, Curtsinger J, Luo X, Waller EK, et al. Human cytomegalovirus (CMV)-induced memory-like NKG2C(+) NK cells are transplantable and expand in vivo in response to recipient CMV antigen. J Immunol 2012;189(10):5082-5088 View Article PubMed/NCBI
  124. Cooper MA, Elliott JM, Keyel PA, Yang L, Carrero JA, Yokoyama WM. Cytokine-induced memory-like natural killer cells. Proc Natl Acad Sci U S A 2009;106(6):1915-1919 View Article PubMed/NCBI
  125. Pahl JHW, Cerwenka A, Ni J. Memory-Like NK Cells: Remembering a Previous Activation by Cytokines and NK Cell Receptors. Front Immunol 2018;9:2796 View Article PubMed/NCBI
  126. Jin F, Lin H, Gao S, Hu Z, Zuo S, Sun L, et al. The anti-tumor role of NK cells in vivo pre-activated and re-stimulated by interleukins in acute lymphoblastic leukemia. Oncotarget 2016;7(48):79187-79202 View Article PubMed/NCBI
  127. Brillantes M, Beaulieu AM. Memory and Memory-Like NK Cell Responses to Microbial Pathogens. Front Cell Infect Microbiol 2020;10:102 View Article PubMed/NCBI
  128. Gang M, Wong P, Berrien-Elliott MM, Fehniger TA. Memory-like natural killer cells for cancer immunotherapy. Semin Hematol 2020;57(4):185-193 View Article PubMed/NCBI
  129. Fehniger TA, Cooper MA. Harnessing NK Cell Memory for Cancer Immunotherapy. Trends Immunol 2016;37(12):877-888 View Article PubMed/NCBI
  130. Tarannum M, Romee R. Cytokine-induced memory-like natural killer cells for cancer immunotherapy. Stem Cell Res Ther 2021;12(1):592 View Article PubMed/NCBI
  131. Galán-Díez M, Cuesta-Domínguez Á, Kousteni S. The Bone Marrow Microenvironment in Health and Myeloid Malignancy. Cold Spring Harb Perspect Med 2018;8(7):a031328 View Article PubMed/NCBI
  132. Boyd AL, Reid JC, Salci KR, Aslostovar L, Benoit YD, Shapovalova Z, et al. Acute myeloid leukaemia disrupts endogenous myelo-erythropoiesis by compromising the adipocyte bone marrow niche. Nat Cell Biol 2017;19(11):1336-1347 View Article PubMed/NCBI
  133. Al-Matary YS, Botezatu L, Opalka B, Hönes JM, Lams RF, Thivakaran A, et al. Acute myeloid leukemia cells polarize macrophages towards a leukemia supporting state in a Growth factor independence 1 dependent manner. Haematologica 2016;101(10):1216-1227 View Article PubMed/NCBI
  134. Lamble AJ, Kosaka Y, Laderas T, Maffit A, Kaempf A, Brady LK, et al. Reversible suppression of T cell function in the bone marrow microenvironment of acute myeloid leukemia. Proc Natl Acad Sci U S A 2020;117(25):14331-14341 View Article PubMed/NCBI
  135. Khaznadar Z, Boissel N, Agaugué S, Henry G, Cheok M, Vignon M, et al. Defective NK Cells in Acute Myeloid Leukemia Patients at Diagnosis Are Associated with Blast Transcriptional Signatures of Immune Evasion. J Immunol 2015;195(6):2580-2590 View Article PubMed/NCBI
  136. Lion E, Willemen Y, Berneman ZN, Van Tendeloo VF, Smits EL. Natural killer cell immune escape in acute myeloid leukemia. Leukemia 2012;26(9):2019-2026 View Article PubMed/NCBI
  137. Guo R, Lü M, Cao F, Wu G, Gao F, Pang H, et al. Single-cell map of diverse immune phenotypes in the acute myeloid leukemia microenvironment. Biomark Res 2021;9(1):15 View Article PubMed/NCBI
  138. Kuznetsova V, Patel S, Luca F, Camacho V, Matkins V, Welner RS. Perturbed function of natural killer cells by inflammatory cytokines in acute (AML) and chronic (CML) myeloid leukemias. J Immunol 2022;208(Suppl 1):62.04 View Article
  139. Rey J, Fauriat C, Kochbati E, Orlanducci F, Charbonnier A, D’Incan E, et al. Kinetics of Cytotoxic Lymphocytes Reconstitution after Induction Chemotherapy in Elderly AML Patients Reveals Progressive Recovery of Normal Phenotypic and Functional Features in NK Cells. Front Immunol 2017;8:64 View Article PubMed/NCBI
  140. Dunbar EM, Buzzeo MP, Levine JB, Schold JD, Meier-Kriesche HU, Reddy V. The relationship between circulating natural killer cells after reduced intensity conditioning hematopoietic stem cell transplantation and relapse-free survival and graft-versus-host disease. Haematologica 2008;93(12):1852-1858 View Article PubMed/NCBI
  141. Kim SY, Lee H, Han MS, Shim H, Eom HS, Park B, et al. Post-Transplantation Natural Killer Cell Count: A Predictor of Acute Graft-Versus-Host Disease and Survival Outcomes After Allogeneic Hematopoietic Stem Cell Transplantation. Clin Lymphoma Myeloma Leuk 2016;16(9):527-535.e2 View Article PubMed/NCBI
  142. Mushtaq MU, Shahzad M, Shah AY, Chaudhary SG, Zafar MU, Anwar I, et al. Impact of natural killer cells on outcomes after allogeneic hematopoietic stem cell transplantation: A systematic review and meta-analysis. Front Immunol 2022;13:1005031 View Article PubMed/NCBI
  143. Lang P, Pfeiffer M, Teltschik HM, Schlegel P, Feuchtinger T, Ebinger M, et al. Natural killer cell activity influences outcome after T cell depleted stem cell transplantation from matched unrelated and haploidentical donors. Best Pract Res Clin Haematol 2011;24(3):403-411 View Article PubMed/NCBI
  144. Jamal E, Azmy E, Ayed M, Aref S, Eisa N. Clinical Impact of Percentage of Natural Killer Cells and Natural Killer-Like T Cell Population in Acute Myeloid Leukemia. J Hematol 2020;9(3):62-70 View Article PubMed/NCBI
  145. Costello RT, Sivori S, Marcenaro E, Lafage-Pochitaloff M, Mozziconacci MJ, Reviron D, et al. Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia. Blood 2002;99(10):3661-3667 View Article PubMed/NCBI
  146. Sivori S, Parolini S, Marcenaro E, Castriconi R, Pende D, Millo R, et al. Involvement of natural cytotoxicity receptors in human natural killer cell-mediated lysis of neuroblastoma and glioblastoma cell lines. J Neuroimmunol 2000;107(2):220-225 View Article PubMed/NCBI
  147. Barrow AD, Martin CJ, Colonna M. The Natural Cytotoxicity Receptors in Health and Disease. Front Immunol 2019;10:909 View Article PubMed/NCBI
  148. Sanchez-Correa B, Morgado S, Gayoso I, Bergua JM, Casado JG, Arcos MJ, et al. Human NK cells in acute myeloid leukaemia patients: analysis of NK cell-activating receptors and their ligands. Cancer Immunol Immunother 2011;60(8):1195-1205 View Article PubMed/NCBI
  149. Fauriat C, Just-Landi S, Mallet F, Arnoulet C, Sainty D, Olive D, et al. Deficient expression of NCR in NK cells from acute myeloid leukemia: Evolution during leukemia treatment and impact of leukemia cells in NCRdull phenotype induction. Blood 2007;109(1):323-330 View Article PubMed/NCBI
  150. Nowbakht P, Ionescu MC, Rohner A, Kalberer CP, Rossy E, Mori L, et al. Ligands for natural killer cell-activating receptors are expressed upon the maturation of normal myelomonocytic cells but at low levels in acute myeloid leukemias. Blood 2005;105(9):3615-3622 View Article PubMed/NCBI
  151. Dhanasekaran R, Deutzmann A, Mahauad-Fernandez WD, Hansen AS, Gouw AM, Felsher DW. The MYC oncogene - the grand orchestrator of cancer growth and immune evasion. Nat Rev Clin Oncol 2022;19(1):23-36 View Article PubMed/NCBI
  152. Nanbakhsh A, Pochon C, Mallavialle A, Amsellem S, Bourhis JH, Chouaib S. c-Myc regulates expression of NKG2D ligands ULBP1/2/3 in AML and modulates their susceptibility to NK-mediated lysis. Blood 2014;123(23):3585-3595 View Article PubMed/NCBI
  153. Wu Z, Zhang H, Wu M, Peng G, He Y, Wan N, et al. Targeting the NKG2D/NKG2D-L axis in acute myeloid leukemia. Biomed Pharmacother 2021;137:111299 View Article PubMed/NCBI
  154. Baragaño Raneros A, Martín-Palanco V, Fernandez AF, Rodriguez RM, Fraga MF, Lopez-Larrea C, et al. Methylation of NKG2D ligands contributes to immune system evasion in acute myeloid leukemia. Genes Immun 2015;16(1):71-82 View Article PubMed/NCBI
  155. Groh V, Wu J, Yee C, Spies T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 2002;419(6908):734-738 View Article PubMed/NCBI
  156. Hilpert J, Grosse-Hovest L, Grünebach F, Buechele C, Nuebling T, Raum T, et al. Comprehensive analysis of NKG2D ligand expression and release in leukemia: implications for NKG2D-mediated NK cell responses. J Immunol 2012;189(3):1360-1371 View Article PubMed/NCBI
  157. Ferrari de Andrade L, Tay RE, Pan D, Luoma AM, Ito Y, Badrinath S, et al. Antibody-mediated inhibition of MICA and MICB shedding promotes NK cell-driven tumor immunity. Science 2018;359(6383):1537-1542 View Article PubMed/NCBI
  158. Ferrari de Andrade L, Kumar S, Luoma AM, Ito Y, Alves da Silva PH, Pan D, et al. Inhibition of MICA and MICB Shedding Elicits NK-Cell-Mediated Immunity against Tumors Resistant to Cytotoxic T Cells. Cancer Immunol Res 2020;8(6):769-780 View Article PubMed/NCBI
  159. Jin Z, Ye W, Lan T, Zhao Y, Liu X, Chen J, et al. Characteristic of TIGIT and DNAM-1 Expression on Foxp3+ γδ T Cells in AML Patients. Biomed Res Int 2020;2020:4612952 View Article PubMed/NCBI
  160. Chashchina A, Märklin M, Hinterleitner C, Salih HR, Heitmann JS, Klimovich B. DNAM-1/CD226 is functionally expressed on acute myeloid leukemia (AML) cells and is associated with favorable prognosis. Sci Rep 2021;11(1):18012 View Article PubMed/NCBI
  161. Paolini R, Molfetta R. Dysregulation of DNAM-1-Mediated NK Cell Anti-Cancer Responses in the Tumor Microenvironment. Cancers (Basel) 2023;15(18):4616 View Article PubMed/NCBI
  162. Stringaris K, Sekine T, Khoder A, Alsuliman A, Razzaghi B, Sargeant R, et al. Leukemia-induced phenotypic and functional defects in natural killer cells predict failure to achieve remission in acute myeloid leukemia. Haematologica 2014;99(5):836-847 View Article PubMed/NCBI
  163. Sandoval-Borrego D, Moreno-Lafont MC, Vazquez-Sanchez EA, Gutierrez-Hoya A, López-Santiago R, Montiel-Cervantes LA, et al. Overexpression of CD158 and NKG2A Inhibitory Receptors and Underexpression of NKG2D and NKp46 Activating Receptors on NK Cells in Acute Myeloid Leukemia. Arch Med Res 2016;47(1):55-64 View Article PubMed/NCBI
  164. Lagana A, Ruan DF, Melnekoff D, Leshchenko V, Perumal D, Rahman A, et al. Increased HLA-E Expression Correlates with Early Relapse in Multiple Myeloma. Blood 2018;132(Suppl 1):59 View Article
  165. Seliger B, Jasinski-Bergner S, Quandt D, Stoehr C, Bukur J, Wach S, et al. HLA-E expression and its clinical relevance in human renal cell carcinoma. Oncotarget 2016;7(41):67360-67372 View Article PubMed/NCBI
  166. Ruggeri L, Urbani E, André P, Mancusi A, Tosti A, Topini F, et al. Effects of anti-NKG2A antibody administration on leukemia and normal hematopoietic cells. Haematologica 2016;101(5):626-633 View Article PubMed/NCBI
  167. Shen M, Linn YC, Ren EC. KIR-HLA profiling shows presence of higher frequencies of strong inhibitory KIR-ligands among prognostically poor risk AML patients. Immunogenetics 2016;68(2):133-144 View Article PubMed/NCBI
  168. Ghasemimehr N, Moazed V, Fatemi A. Gene expression analysis of activating and inhibitory receptors of natural killer cells in patients with acute myeloblastic leukemia. Adv Med Sci 2020;65(2):354-360 View Article PubMed/NCBI
  169. Yang L, Feng Y, Wang S, Jiang S, Tao L, Li J, et al. Siglec-7 is an indicator of natural killer cell function in acute myeloid leukemia. Int Immunopharmacol 2021;99:107965 View Article PubMed/NCBI
  170. Mundy-Bosse BL, Scoville SD, Chen L, McConnell K, Mao HC, Ahmed EH, et al. MicroRNA-29b mediates altered innate immune development in acute leukemia. J Clin Invest 2016;126(12):4404-4416 View Article PubMed/NCBI
  171. Chretien AS, Granjeaud S, Gondois-Rey F, Harbi S, Orlanducci F, Blaise D, et al. Increased NK Cell Maturation in Patients with Acute Myeloid Leukemia. Front Immunol 2015;6:564 View Article PubMed/NCBI
  172. Chretien AS, Fauriat C, Orlanducci F, Galseran C, Rey J, Bouvier Borg G, et al. Natural Killer Defective Maturation Is Associated with Adverse Clinical Outcome in Patients with Acute Myeloid Leukemia. Front Immunol 2017;8:573 View Article PubMed/NCBI
  173. Chretien AS, Devillier R, Granjeaud S, Cordier C, Demerle C, Salem N, et al. High-dimensional mass cytometry analysis of NK cell alterations in AML identifies a subgroup with adverse clinical outcome. Proc Natl Acad Sci U S A 2021;118(22):e2020459118 View Article PubMed/NCBI
  174. Liu L, Chen X, Jin HM, Zhao SS, Zhu Y, Qian SX, et al. [The Expression and Function of NK Cells in Patients with Acute Myeloid Leukemia]. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2022;30(1):49-55 View Article PubMed/NCBI
  175. Crinier A, Dumas PY, Escalière B, Piperoglou C, Gil L, Villacreces A, et al. Single-cell profiling reveals the trajectories of natural killer cell differentiation in bone marrow and a stress signature induced by acute myeloid leukemia. Cell Mol Immunol 2021;18(5):1290-1304 View Article PubMed/NCBI
  176. Shibru B, Fey K, Fricke S, Blaudszun AR, Fürst F, Weise M, et al. Detection of Immune Checkpoint Receptors - A Current Challenge in Clinical Flow Cytometry. Front Immunol 2021;12:694055 View Article PubMed/NCBI
  177. Shiravand Y, Khodadadi F, Kashani SMA, Hosseini-Fard SR, Hosseini S, Sadeghirad H, et al. Immune Checkpoint Inhibitors in Cancer Therapy. Curr Oncol 2022;29(5):3044-3060 View Article PubMed/NCBI
  178. Trefny MP, Kaiser M, Stanczak MA, Herzig P, Savic S, Wiese M, et al. PD-1(+) natural killer cells in human non-small cell lung cancer can be activated by PD-1/PD-L1 blockade. Cancer Immunol Immunother 2020;69(8):1505-1517 View Article PubMed/NCBI
  179. Concha-Benavente F, Kansy B, Moskovitz J, Moy J, Chandran U, Ferris RL. PD-L1 Mediates Dysfunction in Activated PD-1(+) NK Cells in Head and Neck Cancer Patients. Cancer Immunol Res 2018;6(12):1548-1560 View Article PubMed/NCBI
  180. Jimbu L, Mesaros O, Popescu C, Neaga A, Berceanu I, Dima D, et al. Is There a Place for PD-1-PD-L Blockade in Acute Myeloid Leukemia?. Pharmaceuticals (Basel) 2021;14(4):288 View Article PubMed/NCBI
  181. Brodská B, Otevřelová P, Šálek C, Fuchs O, Gašová Z, Kuželová K. High PD-L1 Expression Predicts for Worse Outcome of Leukemia Patients with Concomitant NPM1 and FLT3 Mutations. Int J Mol Sci 2019;20(11):2823 View Article PubMed/NCBI
  182. Wang F, Yang L, Xiao M, Zhang Z, Shen J, Anuchapreeda S, et al. PD-L1 regulates cell proliferation and apoptosis in acute myeloid leukemia by activating PI3K-AKT signaling pathway. Sci Rep 2022;12(1):11444 View Article PubMed/NCBI
  183. Chen C, Liang C, Wang S, Chio CL, Zhang Y, Zeng C, et al. Expression patterns of immune checkpoints in acute myeloid leukemia. J Hematol Oncol 2020;13(1):28 View Article PubMed/NCBI
  184. Darwish NH, Sudha T, Godugu K, Elbaz O, Abdelghaffar HA, Hassan EE, et al. Acute myeloid leukemia stem cell markers in prognosis and targeted therapy: potential impact of BMI-1, TIM-3 and CLL-1. Oncotarget 2016;7(36):57811-57820 View Article PubMed/NCBI
  185. Xu L, Xu J, Ma S, Li X, Zhu M, Chen S, et al. High Tim-3 expression on AML blasts could enhance chemotherapy sensitivity. Oncotarget 2017;8(60):102088-102096 View Article PubMed/NCBI
  186. Kamal AM, Nabih NA, Elleboudy NS, Radwan SM. Expression of immune check point gene TIM-3 in patients newly diagnosed with acute myeloid leukemia: Significance and impact on outcome. Oncol Lett 2021;21(4):325 View Article PubMed/NCBI
  187. Rakova J, Truxova I, Holicek P, Salek C, Hensler M, Kasikova L, et al. TIM-3 levels correlate with enhanced NK cell cytotoxicity and improved clinical outcome in AML patients. Oncoimmunology 2021;10(1):1889822 View Article PubMed/NCBI
  188. Kikushige Y, Miyamoto T, Yuda J, Jabbarzadeh-Tabrizi S, Shima T, Takayanagi S, et al. A TIM-3/Gal-9 Autocrine Stimulatory Loop Drives Self-Renewal of Human Myeloid Leukemia Stem Cells and Leukemic Progression. Cell Stem Cell 2015;17(3):341-352 View Article PubMed/NCBI
  189. Ruggeri L, Capanni M, Casucci M, Volpi I, Tosti A, Perruccio K, et al. Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood 1999;94(1):333-339 PubMed/NCBI
  190. Ruggeri L, Capanni M, Mancusi A, Martelli MF, Velardi A. The impact of donor natural killer cell alloreactivity on allogeneic hematopoietic transplantation. Transpl Immunol 2005;14(3-4):203-206 View Article PubMed/NCBI
  191. Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, Tosti A, et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002;295(5562):2097-2100 View Article PubMed/NCBI
  192. Mancusi A, Ruggeri L, Urbani E, Pierini A, Massei MS, Carotti A, et al. Haploidentical hematopoietic transplantation from KIR ligand-mismatched donors with activating KIRs reduces nonrelapse mortality. Blood 2015;125(20):3173-3182 View Article PubMed/NCBI
  193. Xu J, Niu T. Natural killer cell-based immunotherapy for acute myeloid leukemia. J Hematol Oncol 2020;13(1):167 View Article PubMed/NCBI
  194. Lupo KB, Matosevic S. Natural Killer Cells as Allogeneic Effectors in Adoptive Cancer Immunotherapy. Cancers (Basel) 2019;11(6):769 View Article PubMed/NCBI
  195. Farag SS, Caligiuri MA. Cytokine modulation of the innate immune system in the treatment of leukemia and lymphoma. Adv Pharmacol 2004;51:295-318 View Article PubMed/NCBI
  196. Veluchamy JP, Kok N, van der Vliet HJ, Verheul HMW, de Gruijl TD, Spanholtz J. The Rise of Allogeneic Natural Killer Cells As a Platform for Cancer Immunotherapy: Recent Innovations and Future Developments. Front Immunol 2017;8:631 View Article PubMed/NCBI
  197. Hong G, Xie S, Guo Z, Zhang D, Ge S, Zhang S, et al. Progression-Free Survival of a Patient with Advanced Hepatocellular Carcinoma Treated with Adoptive Cell Therapy Using Natural Killer Cells: A Case Report. Onco Targets Ther 2022;15:255-266 View Article PubMed/NCBI
  198. Nahi H, Chrobok M, Meinke S, Gran C, Marquardt N, Afram G, et al. Autologous NK cells as consolidation therapy following stem cell transplantation in multiple myeloma. Cell Rep Med 2022;3(2):100508 View Article PubMed/NCBI
  199. Wang D, Sun Z, Zhu X, Zheng X, Zhou Y, Lu Y, et al. GARP-mediated active TGF-β1 induces bone marrow NK cell dysfunction in AML patients with early relapse post-allo-HSCT. Blood 2022;140(26):2788-2804 View Article PubMed/NCBI
  200. Sola C, Blery M, Bonnafous C, Bonnet E, Fuseri N, Graziano RF, et al. Lirilumab Enhances Anti-Tumor Efficacy of Elotuzumab. Blood 2014;124(21):4711 View Article
  201. Vey N, Bourhis JH, Boissel N, Bordessoule D, Prebet T, Charbonnier A, et al. A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission. Blood 2012;120(22):4317-4323 View Article PubMed/NCBI
  202. Vey N, Dumas PY, Recher C, Gastaud L, Lioure B, Bulabois CE, et al. Randomized Phase 2 Trial of Lirilumab (anti-KIR monoclonal antibody, mAb) As Maintenance Treatment in Elderly Patients (pts) with Acute Myeloid Leukemia (AML): Results of the Effikir Trial. Blood 2017;130(Suppl 1):889 View Article
  203. Xiao J, Zhang T, Gao F, Zhou Z, Shu G, Zou Y, et al. Natural Killer Cells: A Promising Kit in the Adoptive Cell Therapy Toolbox. Cancers (Basel) 2022;14(22):5657 View Article PubMed/NCBI
  204. Passweg JR, Tichelli A, Meyer-Monard S, Heim D, Stern M, Kühne T, et al. Purified donor NK-lymphocyte infusion to consolidate engraftment after haploidentical stem cell transplantation. Leukemia 2004;18(11):1835-1838 View Article PubMed/NCBI
  205. Stern M, Passweg JR, Meyer-Monard S, Esser R, Tonn T, Soerensen J, et al. Pre-emptive immunotherapy with purified natural killer cells after haploidentical SCT: a prospective phase II study in two centers. Bone Marrow Transplant 2013;48(3):433-438 View Article PubMed/NCBI
  206. Ciurea SO, Schafer JR, Bassett R, Denman CJ, Cao K, Willis D, et al. Phase 1 clinical trial using mbIL21 ex vivo-expanded donor-derived NK cells after haploidentical transplantation. Blood 2017;130(16):1857-1868 View Article PubMed/NCBI
  207. Choi I, Yoon SR, Park SY, Kim H, Jung SJ, Jang YJ, et al. Donor-derived natural killer cells infused after human leukocyte antigen-haploidentical hematopoietic cell transplantation: a dose-escalation study. Biol Blood Marrow Transplant 2014;20(5):696-704 View Article PubMed/NCBI
  208. Shaffer BC, Le Luduec JB, Forlenza C, Jakubowski AA, Perales MA, Young JW, et al. Phase II Study of Haploidentical Natural Killer Cell Infusion for Treatment of Relapsed or Persistent Myeloid Malignancies Following Allogeneic Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2016;22(4):705-709 View Article PubMed/NCBI
  209. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005;105(8):3051-3057 View Article PubMed/NCBI
  210. Lee DA, Denman CJ, Rondon G, Woodworth G, Chen J, Fisher T, et al. Haploidentical Natural Killer Cells Infused before Allogeneic Stem Cell Transplantation for Myeloid Malignancies: A Phase I Trial. Biol Blood Marrow Transplant 2016;22(7):1290-1298 View Article PubMed/NCBI
  211. Bachanova V, Cooley S, Defor TE, Verneris MR, Zhang B, McKenna DH, et al. Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood 2014;123(25):3855-3863 View Article PubMed/NCBI
  212. Mardiana S, Gill S. CAR T Cells for Acute Myeloid Leukemia: State of the Art and Future Directions. Front Oncol 2020;10:697 View Article PubMed/NCBI
  213. Bi X, Hsu J, Gergis M, Yang Y, Yi D, Gergis U. Chimeric Antigen Receptor T-cell Therapy for Acute Myeloid Leukemia. Hematol Oncol Stem Cell Ther 2022;15(3):131-136 View Article PubMed/NCBI
  214. Cummins KD, Gill S. Chimeric antigen receptor T-cell therapy for acute myeloid leukemia: how close to reality?. Haematologica 2019;104(7):1302-1308 View Article PubMed/NCBI
  215. Shah NN, Fry TJ. Mechanisms of resistance to CAR T cell therapy. Nat Rev Clin Oncol 2019;16(6):372-385 View Article PubMed/NCBI
  216. Klingemann H. Are natural killer cells superior CAR drivers?. Oncoimmunology 2014;3:e28147 View Article PubMed/NCBI
  217. Liu E, Marin D, Banerjee P, Macapinlac HA, Thompson P, Basar R, et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N Engl J Med 2020;382(6):545-553 View Article PubMed/NCBI
  218. Caruso S, De Angelis B, Del Bufalo F, Ciccone R, Donsante S, Volpe G, et al. Safe and effective off-the-shelf immunotherapy based on CAR.CD123-NK cells for the treatment of acute myeloid leukaemia. J Hematol Oncol 2022;15(1):163 View Article PubMed/NCBI
  219. Garrison B, Deng H, Yucel G, Frankel NW, Gordley R, Hung M, et al. Senti-202, a Selective, Off-the-Shelf, Preclinical CAR-NK Cell Therapy with CD33 and/or FLT3 Activating CAR, Healthy Cell Protection from Endomucin (EMCN) Inhibitory CAR and Calibrated Release IL-15 for Hematologic Malignancies Including AML. Blood 2022;382(Suppl 1):4531-4532 View Article
  220. Huang R, Wen Q, Wang X, Yan H, Ma Y, Mai-Hong W, et al. Off-the-Shelf CD33 CAR-NK Cell Therapy for Relapse/Refractory AML: First-in-Human, Phase I Trial. Blood 2022;140(Suppl 1):7450-7451 View Article
  221. Ureña-Bailén G, Dobrowolski JM, Hou Y, Dirlam A, Roig-Merino A, Schleicher S, et al. Preclinical Evaluation of CRISPR-Edited CAR-NK-92 Cells for Off-the-Shelf Treatment of AML and B-ALL. Int J Mol Sci 2022;23(21):12828 View Article PubMed/NCBI
  222. Grote S, Ureña-Bailén G, Chan KC, Baden C, Mezger M, Handgretinger R, et al. In Vitro Evaluation of CD276-CAR NK-92 Functionality, Migration and Invasion Potential in the Presence of Immune Inhibitory Factors of the Tumor Microenvironment. Cells 2021;10(5):1020 View Article PubMed/NCBI
  223. ClinicalTrials.gov. Study of Anti-CD33/CLL1 CAR-NK in Acute Myeloid Leukemia. ClinicalTrials.gov identifier: NCT05215015. Available from: https://clinicaltrials.gov/study/NCT05215015. Accessed September 18, 2023
  224. ClinicalTrials.gov. NKG2D CAR-NK Cell Therapy in Patients With Relapsed or Refractory Acute Myeloid Leukemia. ClinicalTrials.gov identifier: NCT05247957. Available from: https://clinicaltrials.gov/study/NCT05247957. Accessed September 18, 2023
  225. Riegg F, Lutz MS, Schmied BJ, Heitmann JS, Queudeville M, Lang P, et al. An Fc-Optimized CD133 Antibody for Induction of NK Cell Reactivity against B Cell Acute Lymphoblastic Leukemia. Cancers (Basel) 2021;13(7):1632 View Article PubMed/NCBI
  226. Koerner SP, André MC, Leibold JS, Kousis PC, Kübler A, Pal M, et al. An Fc-optimized CD133 antibody for induction of NK cell reactivity against myeloid leukemia. Leukemia 2017;31(2):459-469 View Article PubMed/NCBI
  227. Steinbacher J, Baltz-Ghahremanpour K, Schmiedel BJ, Steinle A, Jung G, Kübler A, et al. An Fc-optimized NKG2D-immunoglobulin G fusion protein for induction of natural killer cell reactivity against leukemia. Int J Cancer 2015;136(5):1073-1084 View Article PubMed/NCBI
  228. Castaigne S, Pautas C, Terré C, Raffoux E, Bordessoule D, Bastie JN, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet 2012;379(9825):1508-1516 View Article PubMed/NCBI
  229. Amadori S, Suciu S, Selleslag D, Aversa F, Gaidano G, Musso M, et al. Gemtuzumab Ozogamicin Versus Best Supportive Care in Older Patients With Newly Diagnosed Acute Myeloid Leukemia Unsuitable for Intensive Chemotherapy: Results of the Randomized Phase III EORTC-GIMEMA AML-19 Trial. J Clin Oncol 2016;34(9):972-979 View Article PubMed/NCBI
  230. Domínguez JM, Pérez-Chacón G, Guillén MJ, Muñoz-Alonso MJ, Somovilla-Crespo B, Cibrián D, et al. CD13 as a new tumor target for antibody-drug conjugates: validation with the conjugate MI130110. J Hematol Oncol 2020;13(1):32 View Article PubMed/NCBI
  231. Roas M, Vick B, Kasper MA, Able M, Polzer H, Gerlach M, et al. Targeting FLT3 with a new-generation antibody-drug conjugate in combination with kinase inhibitors for treatment of AML. Blood 2023;141(9):1023-1035 View Article PubMed/NCBI
  232. Zheng B, Yu SF, Del Rosario G, Leong SR, Lee GY, Vij R, et al. An Anti-CLL-1 Antibody-Drug Conjugate for the Treatment of Acute Myeloid Leukemia. Clin Cancer Res 2019;25(4):1358-1368 View Article PubMed/NCBI
  233. Foley B, Felices M, Cichocki F, Cooley S, Verneris MR, Miller JS. The biology of NK cells and their receptors affects clinical outcomes after hematopoietic cell transplantation (HCT). Immunol Rev 2014;258(1):45-63 View Article PubMed/NCBI
  234. Ravandi F, Assi R, Daver N, Benton CB, Kadia T, Thompson PA, et al. Idarubicin, cytarabine, and nivolumab in patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome: a single-arm, phase 2 study. Lancet Haematol 2019;6(9):e480-e488 View Article PubMed/NCBI
  235. Daver N, Garcia-Manero G, Basu S, Boddu PC, Alfayez M, Cortes JE, et al. Efficacy, Safety, and Biomarkers of Response to Azacitidine and Nivolumab in Relapsed/Refractory Acute Myeloid Leukemia: A Nonrandomized, Open-Label, Phase II Study. Cancer Discov 2019;9(3):370-383 View Article PubMed/NCBI
  236. Brunner A, Borate U, Esteve J, Porkka K, Knapper S, Vey N, et al. Aml-190:anti-tim-3 antibody mbg453 in combination with hypomethylating agents (hmas)in patients with high-risk myelodysplastic syndrome (hr-mds) and acute myeloidleukemia: a phase 1 study. Clin Lymphoma Myeloma Leukemia 2020;20(Suppl 1):S188-S189 View Article
  237. Tan J, Tan H, Li Y. Targeting TIM-3 for hematological malignancy: latest updates from the 2022 ASH annual meeting. Exp Hematol Oncol 2023;12(1):62 View Article PubMed/NCBI
  238. Gauthier L, Morel A, Anceriz N, Rossi B, Blanchard-Alvarez A, Grondin G, et al. Multifunctional Natural Killer Cell Engagers Targeting NKp46 Trigger Protective Tumor Immunity. Cell 2019;177(7):1701-1713.e16 View Article PubMed/NCBI
  239. Wang T, Sun F, Xie W, Tang M, He H, Jia X, et al. A bispecific protein rG7S-MICA recruits natural killer cells and enhances NKG2D-mediated immunosurveillance against hepatocellular carcinoma. Cancer Lett 2016;372(2):166-178 View Article PubMed/NCBI
  240. Vallera DA, Zhang B, Gleason MK, Oh S, Weiner LM, Kaufman DS, et al. Heterodimeric bispecific single-chain variable-fragment antibodies against EpCAM and CD16 induce effective antibody-dependent cellular cytotoxicity against human carcinoma cells. Cancer Biother Radiopharm 2013;28(4):274-282 View Article PubMed/NCBI
  241. Wiernik A, Foley B, Zhang B, Verneris MR, Warlick E, Gleason MK, et al. Targeting natural killer cells to acute myeloid leukemia in vitro with a CD16 x 33 bispecific killer cell engager and ADAM17 inhibition. Clin Cancer Res 2013;19(14):3844-3855 View Article PubMed/NCBI
  242. Gleason MK, Ross JA, Warlick ED, Lund TC, Verneris MR, Wiernik A, et al. CD16xCD33 bispecific killer cell engager (BiKE) activates NK cells against primary MDS and MDSC CD33+ targets. Blood 2014;123(19):3016-3026 View Article PubMed/NCBI
  243. Vallera DA, Felices M, McElmurry R, McCullar V, Zhou X, Schmohl JU, et al. IL15 Trispecific Killer Engagers (TriKE) Make Natural Killer Cells Specific to CD33+ Targets While Also Inducing Persistence, In Vivo Expansion, and Enhanced Function. Clin Cancer Res 2016;22(14):3440-3450 View Article PubMed/NCBI
  244. Felices M, Lenvik TR, Kodal B, Lenvik AJ, Hinderlie P, Bendzick LE, et al. Potent Cytolytic Activity and Specific IL15 Delivery in a Second-Generation Trispecific Killer Engager. Cancer Immunol Res 2020;8(9):1139-1149 View Article PubMed/NCBI
  245. Arvindam US, van Hauten PMM, Schirm D, Schaap N, Hobo W, Blazar BR, et al. A trispecific killer engager molecule against CLEC12A effectively induces NK-cell mediated killing of AML cells. Leukemia 2021;35(6):1586-1596 View Article PubMed/NCBI
  246. Goebeler ME, Stuhler G, Bargou R. Bispecific and multispecific antibodies in oncology: opportunities and challenges. Nat Rev Clin Oncol 2024;21:539-560 View Article PubMed/NCBI
  247. Reusing SB, Vallera DA, Manser AR, Vatrin T, Bhatia S, Felices M, et al. CD16xCD33 Bispecific Killer Cell Engager (BiKE) as potential immunotherapeutic in pediatric patients with AML and biphenotypic ALL. Cancer Immunol Immunother 2021;70(12):3701-3708 View Article PubMed/NCBI
  248. Warlick DE, Weisdorf DJ, Vallera DA, Wangen R, Lewis D, Knox J, et al. GTB-3550 TriKE™ for the Treatment of High-Risk Myelodysplastic Syndromes (MDS) and Refractory/Relapsed Acute Myeloid Leukemia (AML) Safely Drives Natural Killer (NK) Cell Proliferation At Initial Dose Cohorts. Blood 2020;136(Suppl 1):7-8 View Article
  249. Alva A, Daniels GA, Wong MK, Kaufman HL, Morse MA, McDermott DF, et al. Contemporary experience with high-dose interleukin-2 therapy and impact on survival in patients with metastatic melanoma and metastatic renal cell carcinoma. Cancer Immunol Immunother 2016;65(12):1533-1544 View Article PubMed/NCBI
  250. Decot V, Voillard L, Latger-Cannard V, Aissi-Rothé L, Perrier P, Stoltz JF, et al. Natural-killer cell amplification for adoptive leukemia relapse immunotherapy: comparison of three cytokines, IL-2, IL-15, or IL-7 and impact on NKG2D, KIR2DL1, and KIR2DL2 expression. Exp Hematol 2010;38(5):351-362 View Article PubMed/NCBI
  251. Sanchez-Correa B, Bergua JM, Pera A, Campos C, Arcos MJ, Bañas H, et al. In Vitro Culture with Interleukin-15 Leads to Expression of Activating Receptors and Recovery of Natural Killer Cell Function in Acute Myeloid Leukemia Patients. Front Immunol 2017;8:931 View Article PubMed/NCBI
  252. Baer MR, George SL, Caligiuri MA, Sanford BL, Bothun SM, Mrózek K, et al. Low-dose interleukin-2 immunotherapy does not improve outcome of patients age 60 years and older with acute myeloid leukemia in first complete remission: Cancer and Leukemia Group B Study 9720. J Clin Oncol 2008;26(30):4934-4939 View Article PubMed/NCBI
  253. Buyse M, Squifflet P, Lange BJ, Alonzo TA, Larson RA, Kolitz JE, et al. Individual patient data meta-analysis of randomized trials evaluating IL-2 monotherapy as remission maintenance therapy in acute myeloid leukemia. Blood 2011;117(26):7007-7013 View Article PubMed/NCBI
  254. Mi R, Chen L, Wang X, Yin Q, Wang Z, Ma X, et al. A retrospective study on effectiveness of combined recombinant human interferon-α-1b, interleukin-2, and thalidomide for the treatment of acute myeloid leukemia in various disease states. Ann Transl Med 2022;10(24):1382 View Article PubMed/NCBI
  255. Nilsson MS, Hallner A, Brune M, Nilsson S, Thorén FB, Martner A, et al. Immunotherapy with HDC/IL-2 may be clinically efficacious in acute myeloid leukemia of normal karyotype. Hum Vaccin Immunother 2020;16(1):109-111 View Article PubMed/NCBI
  256. Zeng Q, Xiang B, Liu Z. Autologous hematopoietic stem cell transplantation followed by interleukin-2 for adult acute myeloid leukemia patients with favorable or intermediate risk after complete remission. Ann Hematol 2022;101(8):1711-1718 View Article PubMed/NCBI
  257. Petit A, Ducassou S, Leblanc T, Pasquet M, Rousseau A, Ragu C, et al. Maintenance Therapy With Interleukin-2 for Childhood AML: Results of ELAM02 Phase III Randomized Trial. Hemasphere 2018;2(6):e159 View Article PubMed/NCBI
  258. Dubois SP, Miljkovic MD, Fleisher TA, Pittaluga S, Hsu-Albert J, Bryant BR, et al. Short-course IL-15 given as a continuous infusion led to a massive expansion of effective NK cells: implications for combination therapy with antitumor antibodies. J Immunother Cancer 2021;9(4):e002193 View Article PubMed/NCBI
  259. Conlon KC, Potter EL, Pittaluga S, Lee CR, Miljkovic MD, Fleisher TA, et al. IL15 by Continuous Intravenous Infusion to Adult Patients with Solid Tumors in a Phase I Trial Induced Dramatic NK-Cell Subset Expansion. Clin Cancer Res 2019;25(16):4945-4954 View Article PubMed/NCBI
  260. Xiong Y, Bensoussan D, Decot V. IL-15 as a potential target in leukemia. Blood Lymphat Cancer 2015;5:55-63 View Article
  261. Cario G, Izraeli S, Teichert A, Rhein P, Skokowa J, Möricke A, et al. High interleukin-15 expression characterizes childhood acute lymphoblastic leukemia with involvement of the CNS. J Clin Oncol 2007;25(30):4813-4820 View Article PubMed/NCBI
  262. Williams MT, Yousafzai Y, Cox C, Blair A, Carmody R, Sai S, et al. Interleukin-15 enhances cellular proliferation and upregulates CNS homing molecules in pre-B acute lymphoblastic leukemia. Blood 2014;123(20):3116-3127 View Article PubMed/NCBI
  263. Petranovic D, Pilcic G, Valkovic T, Sotosek Tokmadzic V, Laskarin G. Perforin- and granulysin-mediated cytotoxicity and interleukin 15 play roles in neurocognitive impairment in patients with acute lymphoblastic leukaemia. Med Hypotheses 2014;83(1):122-126 View Article PubMed/NCBI
  264. Szczepanski MJ, Szajnik M, Welsh A, Foon KA, Whiteside TL, Boyiadzis M. Interleukin-15 enhances natural killer cell cytotoxicity in patients with acute myeloid leukemia by upregulating the activating NK cell receptors. Cancer Immunol Immunother 2010;59(1):73-79 View Article PubMed/NCBI
  265. Romee R, Cooley S, Berrien-Elliott MM, Westervelt P, Verneris MR, Wagner JE, et al. First-in-human phase 1 clinical study of the IL-15 superagonist complex ALT-803 to treat relapse after transplantation. Blood 2018;131(23):2515-2527 View Article PubMed/NCBI
  266. Margolin K, Morishima C, Velcheti V, Miller JS, Lee SM, Silk AW, et al. Phase I Trial of ALT-803, A Novel Recombinant IL15 Complex, in Patients with Advanced Solid Tumors. Clin Cancer Res 2018;24(22):5552-5561 View Article PubMed/NCBI
  267. Berrien-Elliott MM, Becker-Hapak M, Cashen AF, Jacobs M, Wong P, Foster M, et al. Systemic IL-15 promotes allogeneic cell rejection in patients treated with natural killer cell adoptive therapy. Blood 2022;139(8):1177-1183 View Article PubMed/NCBI
  268. Du Z, Ng YY, Zha S, Wang S. piggyBac system to co-express NKG2D CAR and IL-15 to augment the in vivo persistence and anti-AML activity of human peripheral blood NK cells. Mol Ther Methods Clin Dev 2021;23:582-596 View Article PubMed/NCBI
  269. Zhao XY, Jiang Q, Jiang H, Hu LJ, Zhao T, Yu XX, et al. Expanded clinical-grade membrane-bound IL-21/4-1BBL NK cell products exhibit activity against acute myeloid leukemia in vivo. Eur J Immunol 2020;50(9):1374-1385 View Article PubMed/NCBI
  270. Rubino V. S254: CD4+ T CELL-DERIVED IL21 REGULATES STEM CELL FATE IN ACUTE MYELOID LEUKEMIA. HemaSphere 2022;6(S3):155-156 View Article
  271. Vasu S, Sharma N, Odonnell L, Bosse K, Lee DA. A phase I clinical trial testing the safety of IL-21-expanded, off-the-shelf, natural killer cells for relapsed/refractory acute myeloid leukemia and myelodysplastic syndrome. J Clin Oncol 2020;38(Suppl 15):TPS7562 View Article
  272. ClinicalTrials.gov. Interleukin-21 (IL-21)- Expanded Natural Killer Cells for Induction of Acute Myeloid Leukemia. ClinicalTrials.gov identifier: NCT02809092. Available from: https://clinicaltrials.gov/study/NCT02809092. Accessed September 22, 2023
  273. Romee R, Rosario M, Berrien-Elliott MM, Wagner JA, Jewell BA, Schappe T, et al. Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia. Sci Transl Med 2016;8(357):357ra123 View Article PubMed/NCBI
  274. Bednarski JJ, Zimmerman C, Cashen AF, Desai S, Foster M, Schappe T, et al. Adoptively Transferred Donor-Derived Cytokine Induced Memory-like NK Cells Persist and Induce Remission in Pediatric Patient with Relapsed Acute Myeloid Leukemia after Hematopoietic Cell Transplantation. Blood 2019;134(Suppl 1):3307 View Article
  275. Bednarski JJ, Zimmerman C, Berrien-Elliott MM, Foltz JA, Becker-Hapak M, Neal CC, et al. Donor memory-like NK cells persist and induce remissions in pediatric patients with relapsed AML after transplant. Blood 2022;139(11):1670-1683 View Article PubMed/NCBI
  276. Murugan D, Murugesan V, Panchapakesan B, Rangasamy L. Nanoparticle Enhancement of Natural Killer (NK) Cell-Based Immunotherapy. Cancers (Basel) 2022;14(21):5438 View Article PubMed/NCBI
  277. Dölen Y, Kreutz M, Gileadi U, Tel J, Vasaturo A, van Dinther EA, et al. Co-delivery of PLGA encapsulated invariant NKT cell agonist with antigenic protein induce strong T cell-mediated antitumor immune responses. Oncoimmunology 2016;5(1):e1068493 View Article PubMed/NCBI
  278. Sanz-Ortega L, Rojas JM, Portilla Y, Pérez-Yagüe S, Barber DF. Magnetic Nanoparticles Attached to the NK Cell Surface for Tumor Targeting in Adoptive Transfer Therapies Does Not Affect Cellular Effector Functions. Front Immunol 2019;10:2073 View Article PubMed/NCBI
  279. Gao S, Li T, Guo Y, Sun C, Xianyu B, Xu H. Selenium-Containing Nanoparticles Combine the NK Cells Mediated Immunotherapy with Radiotherapy and Chemotherapy. Adv Mater 2020;32(12):e1907568 View Article PubMed/NCBI
  280. Au KM, Park SI, Wang AZ. Trispecific natural killer cell nanoengagers for targeted chemoimmunotherapy. Sci Adv 2020;6(27):eaba8564 View Article PubMed/NCBI
  281. Alhallak K, Sun J, Muz B, Jeske A, Yavner J, Bash H, et al. Nanoparticle T cell engagers for the treatment of acute myeloid leukemia. Oncotarget 2021;12(19):1878-1885 View Article PubMed/NCBI
  282. Alizadeh Zeinabad H, Yeoh WJ, Arif M, Lomora M, Banz Y, Riether C, et al. Natural killer cell-mimic nanoparticles can actively target and kill acute myeloid leukemia cells. Biomaterials 2023;298:122126 View Article PubMed/NCBI
  283. Ho KW, Chen IU, Cheng YA, Liao TY, Liu ES, Chen HJ, et al. Double attack strategy for leukemia using a pre-targeting bispecific antibody (CD20 Ab-mPEG scFv) and actively attracting PEGylated liposomal doxorubicin to enhance anti-tumor activity. J Nanobiotechnology 2021;19(1):16 View Article PubMed/NCBI
  284. Marin D, Li Y, Basar R, Rafei H, Daher M, Dou J, et al. Safety, efficacy and determinants of response of allogeneic CD19-specific CAR-NK cells in CD19(+) B cell tumors: a phase 1/2 trial. Nat Med 2024;30(3):772-784 View Article PubMed/NCBI
  • Oncology Advances
  • eISSN 2996-3427
Back to Top

Natural Killer Cell Cellular-based Therapeutic Options to Manage Acute Myeloid Leukemia: Prospects and Challenges

Ogochukwu O. Izuegbuna
  • Reset Zoom
  • Download TIFF